You are on page 1of 374

METABOLOMICS

Edited by Ute Roessner


 
   
 
 
 
 
 
 
 
 
Metabolomics
Edited by Ute Roessner

Published by InTech
Janeza Trdine 9, 51000 Rijeka, Croatia

Copyright © 2012 InTech


All chapters are Open Access distributed under the Creative Commons Attribution 3.0
license, which allows users to download, copy and build upon published articles even for
commercial purposes, as long as the author and publisher are properly credited, which
ensures maximum dissemination and a wider impact of our publications. After this work
has been published by InTech, authors have the right to republish it, in whole or part, in
any publication of which they are the author, and to make other personal use of the
work. Any republication, referencing or personal use of the work must explicitly identify
the original source.

As for readers, this license allows users to download, copy and build upon published
chapters even for commercial purposes, as long as the author and publisher are properly
credited, which ensures maximum dissemination and a wider impact of our publications.

Notice
Statements and opinions expressed in the chapters are these of the individual contributors
and not necessarily those of the editors or publisher. No responsibility is accepted for the
accuracy of information contained in the published chapters. The publisher assumes no
responsibility for any damage or injury to persons or property arising out of the use of any
materials, instructions, methods or ideas contained in the book.

Publishing Process Manager Igor Babic


Technical Editor Teodora Smiljanic
Cover Designer InTech Design Team

First published February, 2012


Printed in Croatia

A free online edition of this book is available at www.intechopen.com


Additional hard copies can be obtained from orders@intechweb.org

Metabolomics, Edited by Ute Roessner


p. cm.
978-953-51-0046-1
 
 

Contents
 

Preface IX

Part 1 Metabolomics of Microbes and Cell Cultures 1

Chapter 1 Metabolomics and Mammalian Cell Culture 3


Kathya De la Luz-Hdez

Chapter 2 Quantitative Metabolomics and


Its Application in Metabolic Engineering of
Microbial Cell Factories Exemplified by the Baker’s Yeast 19
Mario Klimacek

Part 2 Data Analysis and Integration 45

Chapter 3 Online Metabolomics


Databases and Pipelines 47
Adam J. Carroll

Chapter 4 Generic Software Frameworks


for GC-MS Based Metabolomics 73
Nils Hoffmann and Jens Stoye

Chapter 5 Computational Methods to


Interpret and Integrate Metabolomic Data 99
Feng Li, Jiangxin Wang,
Lei Nie and Weiwen Zhang

Chapter 6 Metabotype Concept: Flexibility, Usefulness and


Meaning in Different Biological Populations 131
Nabil Semmar

Chapter 7 Software Techniques for Enabling


High-Throughput Analysis of Metabolomic Datasets 167
Corey D. DeHaven, Anne M. Evans,
Hongping Dai and Kay A. Lawton
VI Contents

Part 3 Metabolomics to Identify


Health Promoting Factors and New Bioactives 193

Chapter 8 Metabolic Pathways as Targets for Drug Screening 195


Wai-Nang Paul Lee, Laszlo G. Boros and Vay-Liang W. Go

Part 4 Metabolomics in Plant Research 211

Chapter 9 New Opportunities in Metabolomics and


Biochemical Phenotyping for Plant Systems Biology 213
Gibon Yves, Rolin Dominique, Deborde Catherine,
Bernillon Stéphane and Moing Annick

Chapter 10 Metabolomics of Endophytic Fungi


Producing Associated Plant Secondary
Metabolites: Progress, Challenges and Opportunities 241
Souvik Kusari and Michael Spiteller

Part 5 Metabolomics in Human Disease Research 267

Chapter 11 Metabolomics in the Analysis of Inflammatory Diseases 269


Sabrina Kapoor, Martin Fitzpatrick, Elizabeth Clay,
Rachel Bayley, Graham R. Wallace and Stephen P. Young

Chapter 12 Clinical Implementation of Metabolomics 289


Akira Imaizumi, Natsumi Nishikata, Hiroo Yoshida, Junya Yoneda,
Shunji Takahena, Mitsuo Takahashi, Toshihiko Ando,
Hiroshi Miyano, Kenji Nagao, Yasushi Noguchi,
Nobuhisa Shimba and Takeshi Kimura

Part 6 Improving Analytics 315

Chapter 13 Improvement in the Number of Analytic Features Detected


by Non-Targeted Metabolomic Analysis: Influence of the
Chromatographic System and the Ionization Technique 317
R. Pandher, E. Naegele, S.M. Fischer and F.I Raynaud

Part 7 Metabolomics in Safety Assessments 329

Chapter 14 Challenges for Metabolomics


as a Tool in Safety Assessments 331
George G. Harrigan and Bruce Chassy

Chapter 15 Metabolomics Approach for Hazard Identification in


Human Health Assessment of Environmental Chemicals 349
Suryanarayana V. Vulimiri, Brian Pachkowski,
Ambuja S. Bale and Babasaheb Sonawane
 

Preface
 

Metabolomics is a new scientific field which has developed with an accelerating speed
over the last decade as demonstrated through the increasing numbers of publications
in scientific journals of any biological research field. These developments are mainly
driven by increasingly robust and sensitive analytical instrumentations allowing the
analysis and quantification of thousands of metabolites from any biological system.
Together with the application of sophisticated computational methodology and
statistical approaches the vast amount of data generated from instrumentation can be
analysed and mined aiding biological and biochemical interpretation. Especially, once
experimental metabolomics data can be integrated with other ‘omics type data such as
from genomics and proteomics analyses the path is paved for a better holistic
understanding of the biological system under investigation.

This book will provide the reader with summaries of the state-of-the-art of the
technologies and methodologies, especially in the data analysis and interpretation
approaches as well as gives insights into exciting applications of metabolomics in
human health studies, safety assessments and plant and microbial research.

 
Dr. Ute Roessner
School of Botany, The University of Melbourne, Victoria,
Australia
Part 1

Metabolomics of Microbes and Cell Cultures


1

Metabolomics and Mammalian Cell Culture


Kathya De la Luz-Hdez
Center of Molecular Immunology
Cuba

1. Introduction
Since the mid-1950s, when pioneering work of Earle and colleagues (1954) enable routine
cell culture, mammalian cell culture has been used in the large-scale production of
recombinant protein and monoclonal antibodies. Mammalian cell lines are preferred as
production host for many pharmaceuticals, since complex post-translational modifications
of the produced proteins (especially glycosylation) are generally not properly performed by
microbial systems (Lake-Ee Quek et al., 2010).
Wagburg described that under batch conditions, mammalian cells display an inefficient
metabolic phenotype characterized by high rates of glucose to lactate conversion (Warburg,
1956) together with partial oxidation of glutamine to ammonia and non-essential amino
acids (Fitzpatrick et al., 1993; Jenkins et al., 1992; Ljunggren and Haggstrom, 1992; Ozturk
and Palsson, 1991). The accumulation of fermentation by-products causes a reduction of the
culture density and product titer that can be realized (Martinelle et al., 1998).
In order to increase the cell productivity a common optimization approach is to grow cells
to moderately high density in fed-batch and the deliberately induce a prolonged, productive
stationary phase. While optimization of this perturbed batch strategy is responsible for the
increase of monoclonal antibodies titer seen over the past decades it has a number of short-
comings, including:
a. the strategy has to be refined for each new cell line,
b. the ultimate metabolic phenotype during prolonged stationary phase varies between
cell lines and it is not always possible to achieve the most productive phenotypes for a
given strain
c. volumetric productivity remains relatively low due to moderate cell density (Lake Ee
Quek et al., 2010).
Other approaches are related with the changes of cell phenotypes through metabolic
engineering or change of culture media conditions in order to manipulate the cellular
metabolic behavior.
Although transcriptomics and proteomics have been explored extensively for mammalian
cell engineering (Korke et al., 2002; Seow et al., 2001; Seth et al., 2007; Smales et al., 2004; de
la Luz et al., 2007, 2008) these tools fall short of generating direct measurements of the
physiological state of the cell. It is essential to combine these techniques with metabolic flux
4 Metabolomics

analysis (MFA) a powerful method to quantify the manifestation of a phenotype: the


intracellular reaction rates or the fluxome. One of the relatively new “omic” sciences is the
field of metabolomics. The metabolome was first described by Oliver and collagues (1998) as
being the set of all of low-molecular-mass compounds synthesized by an organism.
Metabolomics is therefore the analysis of small molecules that constitute the metabolism,
and it offers the closest direct measurements of a cell´s physiological activity (Beecher, 2002;
Khoo and Al-Rubeai, 2007). The metabolomic analysis can be considered as “the
measurement of the change in the relative concentrations of metabolites as the result of the
deletion or overexpression of a gene, should allow the target of a novel gene product to be
located on the metabolic map”. Another definition of the metabolome states that it consists
of “only those native small molecules that are participant’s in general metabolic reactions
and that are required for the maintenance, growth and normal function of a cell” (Khoo and
Al-Rubeai, 2007).
The metabolomic as a new powerful tool to understand the complex processes of large scale
mammalian cell cultures for biopharmaceutical production has not been yet embraced during
process development and scale-up. This is mostly because metabolites are now not the
primary focus and the relationship between metabolites and protein production in different
media are not fully understood. It is for this reason that metabolomics can bridge the gap of
understanding as to the dynamics of metabolism, cell growth and protein production (Khoo
and Al-Rubeai, 2007). The metabolomics can be use to optimize conditions of bioreactors or the
development chemically defined media. Characterizing cell lines, culture media and selection
of cell lines are a vital step in the process development of biologics. In this chapter, we describe
the state-of-the-art of the use of metabolomics tool in mammalian cell lines.

2. Complexity of metabolome analysis


Metabolomics requires the unbiased identification and quantification of all of the
metabolites present in a specific biological sample (from an organism or in vitro).
Metabolites are generally labile species, by their nature are chemically very diverse, and
often present in a wide dynamic range. For analysis of mRNA and proteins one “only”
needs to know the genome sequence of the organism and exploit this information using
nucleic acid hybridization or protein separation followed by MS (although PTM are
problematic). However, the analysis of metabolites is not as straightforward. In contrast to
transcripts or protein identification, metabolites are not organism specific (that is to say,
sequence dependent) (Hollywood et al., 2006).
In addition, their diverse chemical properties make complete metabolite analysis difficult.
Genes are composed of a linear four-letter code, whereas proteins have a 20-letters code of
primary amino acids. Metabolites do not have any fixed codes, and thus a general method of
characterization is difficult. Present methods use the specific chemical properties of these
entities to separate, identify and decipher their structures. Combinatorial approaches allow
for a greater coverage. An ideal metabolic analysis should provide:
a. Give an instantaneous snapshot of all metabolites in any given system,
b. Use analytical methods that have high recovery, experimental robustness,
reproducibility, high resolving power and high sensitivity (Fiehn, 2001) whilst being
able to be applied universally,
Metabolomics and Mammalian Cell Culture 5

c. Provide the unambiguous quantification and identification of metabolites and


(d) factors to be highlighted while easily being incorporated into biochemical network
models (Soo H and Al-Rubeai M, 2007).
Before any metabolome measurements are taken it is essential that metabolism is stopped as
quickly as possible, especially because the enzymes are active. For animal cells liquid N2 is
used to snap freeze the sample, followed by mechanical disruption in order to release the
metabolites (Viant et al., 2005). The next stage of the analysis is to extract the metabolites.
There are many different methods (Tweeddale et al., 1998; Buchholz et al., 2001; Villas-Boas
et al., 2005) and the most common ones are:
- Acid extraction using perchloric acid, followed by freeze thawing, then neutralization
with potassium hydroxide
- Alkali extraction typically using sodium hydroxide, followed by heating (80oC)
- Ethanolic extraction by boiling the sampling in ethanol at 80oC
When the extract is finally ready, the choice of the analytical tool is based on the level of
chemical information required about the metabolites, remembering that there will be a
chemical bias with respect to that method, and the speed of analysis is also another
consideration. The figure 1 shows different methods and approaches used for the
metabolomic analysis.

Fig. 1. Technologies for metabolome analysis. GC-MS: gas chromatography mass


spectrometry, GCxGC-MS: 2 dimensional GC coupled to mass spectrometry, LC-MS: liquid
chromatography mass spectrometry, HPLC: high performance liquid chromatography, LC-
NMR: liquid chromatography coupled to nuclear magnetic resonance, NMR: nuclear
magnetic resonance, LDI-MS: laser desorption ionization mass spectrometry, FT-IR: Fourier
transform infrared spectroscopy
6 Metabolomics

The most popular approaches are:


1. Metabolite target analysis: which is an approach that is restricted to metabolites for
example a particular enzyme system that would be directly affected by any
perturbation
2. Metabolite profiling: which is focused on a specific group of metabolites (for example
lipids) or those associated with a specific pathway; within clinical and pharmaceutical
analysis, this is often called metabolic profiling, which is used to trace the fate of a drug
or metabolite
3. Metabolomics: is the comprehensive analysis of the entire metabolome, under a given
set of conditions
4. Metabonomics: which seeks to measure the fingerprint of biochemical perturbations
caused by disease, drug and toxins.
5. Metabolic fingerprinting: is used to classify samples based on provenance of either their
biological relevance or origin by using a fingerprinting technology that is rapid but
does not necessarily give specific metabolite information.

3. Experimental design
3.1 Cell culture growth, stimulation
The differences in optimized cell culture growth conditions present another major concern
for cell line metabolomics. This is particularly an issue is studies involving comparative
analysis of several different cell types, all of which might contain different levels of glucose,
glutamine and lactate, as well as other nutrients and additives, which will probably lead to
differences in the metabolome of the cells. If possible, it is recommended to use the same
growth medium for all cell lines in the study to reduce variance in metabolic profile that can
be caused by the medium (Cuperlovic-Culf et al., 2010). The standard enhancement of cell
culture medium with serum of animal origin can add another level of complexity in cell
growth condition optimization. Variations in serum can lead to contamination with
exogenous metabolites and alterations of endogenous cell metabolite.
In order to minimize the influence of different cell culture conditions in the metabolomic
final results, proper experimental designs are crucial. Nonetheless, more effort is required in
the future for the determination of metabolic differences caused by various growth
conditions, cell culture age and/or passage number for different cell lines.

3.2 Sample preparation and metabolite extraction


The goal of metabolomics is to analyze all or, at least, as many as possible different
metabolites without selectivity for any particular molecular type and/or characteristics. The
correct sample preparation is the first step in order to ensure the detection of a large number
of metabolites. As metabolic processes may be rapid, varying from milliseconds to minutes
(Gerdtzen et al., 2004; Taoka and Banerjee, 2002), the first necessary step is to rapidly stop
any inherent enzymatic activity or any changes in the metabolite levels. The time and
method of sampling are important issues to be considered to ensure reproducibility in the
analytical sample, especially since a large number of biological replicates is commonly used
(Khoo and Al-Rubeai, 2007).
Metabolomics and Mammalian Cell Culture 7

These methods include freeze-clamping (with lower-temperature receptacles), immediate


freezing in liquid nitrogen or by acidic treatments (ap Rees and Hill, 1994). Freezing in
liquid nitrogen is generally considered to be the easiest way of stopping enzyme activity
provided that cells or tissues are not allowed to partially thaw before extracting metabolites.
In order to prevent this from happening, enzyme activity is inhibited by freezing-drying or
by immediate addition of organic solvents while applying heat. Cells are subsequently
disrupted, releasing the metabolites. Frozen samples may be ground down by sonication,
homogenization by mechanical means in pre-chilled holders (Fiehn et al., 2000) or directly in
an extraction solvent (Orth et al., 1999). The mixture of cell debris, protein, nucleotides and
the desired metabolites need to be separated; this can be done by centrifugation or filtration.
For the complete analysis of a cell culture, it is important to measure both extracellular
(footprint) and intracellular (fingerprint) metabolic profiles. Metabolic footprinting is
technically simple because it requires only centrifugation to separate culture media and cells
before the analysis. Metabolic fingerprinting, although much more technically challenging
because it requires metabolite extraction from cells, provides more complete information about
cellular metabolic processes (Cuperlovic-Culf et al., 2010). Recently a study related with
different metabolite extraction protocols for mammalian cell culture was published (Dietmair
et al., 2010). In this study, the authors compared 12 different extraction methods, according to
their results; extraction in cold 50% aqueous acetonitrile was superior to other methods.

3.3 Analytical Instruments platforms


Currently, the main analytical techniques used for the analysis of the metabolome are
nuclear magnetic resonance spectroscopy (NMR) and hyphenated techniques such as gas
chromatography (GC) and liquid chromatography (LC) coupled to mass spectrometry (MS).
In addition other combinations are possible, e.g. capillary electrophoresis (CE) coupled to
MS or LC coupled to electrochemical detection. Alternatively, Fourier transform infrared
spectroscopy (FTIR) and direct infusion mass spectrometry (DIMS) have been applied
(Dunn and Ellis, 2005; van Greef et al., 2004; Lindon et al., 2007; Koek et al., 2010) without
any prior separation, except for eventual sample preparation. NMR, FTIR and DIMS are
high throughput methods and require minimal sample preparation and may be preferred
techniques for metabolic fingerprint. However, the obtained spectra are composed of the
signals of very many metabolites and elucidation of these complex spectra can be very
complicated. In addition, detection limits for NMR and FTIR are much higher that for MS-
based techniques, limiting the application range to metabolites present in higher
concentrations. Therefore, GC, LC and CE coupled to MS are generally preferred in
metabolomics to allow quantification and identification of as many as possible metabolites.
The general requirements for metabolomic instruments are:
- Excellent sensitivity and resolution for a wide range of molecules types
- The ability to handle a large range of concentrations (from pM to mM) for different
molecular types
- The ability to identify and quantify different molecules
- Short analysis time
- To enable the measurement of many samples without sample degradation during the
measurement
- Reproducible measurement across different centers and time
8 Metabolomics

Several reviews have dealt with the application of NMR and MS in metabolomics (Ala-
Korpela, 2008; Detmer et al., 2007; Griffin, 2003). NMR is a non-invasive, non-destructive,
highly discriminatory and fast method that can analyze rather crude samples. NMR
spectroscopy can be performed without extensive sample preprocessing and separation and
provides several different experimental protocols optimized for mixture analysis and
molecular formula or structure determination. The results of NMR measurements have
proven highly replicable across centers and instruments (Viant et al., 2005). NMR can
provide measurements for different types and sizes of both polar and non-polar molecules
through analysis of different spectral windows. In addition, NMR instruments are highly
versatile and with only minor changes in probes, users can obtain spectral information for
different nuclei (1H, 13C, 15N, and 32P among others) in solvent or solid samples and even in
vivo (Griffin, 2003). NMR is also the only method used in metabolomics that currently
enables direct measurements of molecular diffusion, interactions and chemical exchange.
Several databases and methods are being developed that enable metabolite identification
and quantification from NMR spectra (Table 1). The major problem with NMR technology
as applied to metabolomics is its low sensitivity, which limits the majority of currently
available instruments to measurement of fewer than 100 metabolites.
The role of MS in metabolomic research is constantly expanding, whether the focus is on
profiling (targeted analysis) or pattern-based analysis (Hollywood et al., 2006). Recent
technological advances in separation science, ion sources and mass analyzers have
considerably increased the sensitivity, selectivity, specificity and speed of metabolite
detection and identification by MS. There are five important considerations that need to be
dealt with in any global metabolite analysis by MS:
1. The efficient and unbiased extraction of metabolites from the sample matrix
2. Separation or fractionation of the analytes by chromatography
3. Ionization of the analyte metabolite
4. Detection of mass signals
5. Analyte identification

Name and availability Instrument Additional information


Human Metabolome Project Biological data; chemical and
NMR, MS
(http:/www.hmdb.ca) clinical data specific to humans
Database search for NMR peaks
BMRD (http:/www.bmrb.wisc.edu) NMR
assignment
Prime (http:/prime.psc.riken.jp) MS, NMR
Glom metabolome database
MS Specific to plants
(http:/csbdb.mpimp-glom.mpg.de)
METLIN metabolite database Drug and drug metabolites;
MS
(http:/metlin.scrpps.edu) specific to humans
NIST chemistry WebBook
NMR, MS, IR
(http:/WebBook.nist.gov/chemistry)
Madison metabolomics database
MS, NMR
(http:/mmcd.nmrfarm.wisc.edu)
NMR Lab of biomolecules Databse search for NMR peaks
NMR
(http:/spinportal.magnet.fsu.edu) assigment

Table 1. Databases of metabolomic standard data for quantification and assignment


Metabolomics and Mammalian Cell Culture 9

Separation of analytes before MS detection is an important step leading to detection of


more features, effectively increasing the overall “peak capacity” of the analytical
platform. Separation methods include condensed-phase separation methods and gas-
phase analyte separation. DIMS relies solely on the mass spectrometer to perform
separation and offers an advantage in terms of speed and sample throughput. The
number of identified features in a MS measurements can also be increased by changing
the polarity of the ion source. Positive ion mode electrospray is optimal for basic
metabolites (e.g. amines). Negative ion mode provides optimal measurement for acidic
metabolites. Knowledge of the empirical formula based on exact mass can often be used
to assign one or a few putative identifications that can then be used for searching
metabolic or chemical databases (Table 1).
A comparative outline of the characteristics of NMR and MS methodologies as applied to
metabolomics is provided in Table 2. The two methods are highly compatible and, thus
ideal approach is to combine the results from NMR and MS measurements.
The most common forms of chromatography are GC and LC. GC runs are relative long, at
about 60 min or more (Gummer et al., 2009); however, deconvolution software allows for
the decrease in run times. In LC there is a shift from standard HPLC to UPLC (ultra-
performance liquid chromatography), which can significantly increase resolution sensitivity
and peak capacity (Gummer et al., 2009) due to the reduced particle size, while decreasing
sample volumes and mobile phases. UPLC systems operate at high operating pressures and
use sub-2-µm porous packing. Unlike pressured systems such as LC, CE (capillary
electrophoresis) makes use of an electric field to move molecules towards the detector, much
like gel electrophoresis. CE coupled with UV or LIF (laser-induced fluorescence) detectors
are highly sensitive, but lack selectivity.
GC coupled to MS is one of the most common instrument platforms to be used in
metabolomics experiments. GC-MS instruments using linear quadrupole analysers have
been available for decades providing a robust technology that is amenable to
automation. The identification of a wide range of primary metabolites (often after
derivitisation) is greatly facilitated by the high resolution of capillary GC, the
reproducible fragmentation of metabolites in the mass spectrometer and the ready
availability of large mass spectral libraries (Roessner et al., 2001). Recent developments
in GC-MS have resulted in improvements in both the GC and MS capabilities of this
platform and a move towards the use of high mass accuracy/ high mass resolution
instruments. The requirement for high throughput has led to the use of nominal GC-
time-of-flight TOF-MS with much faster scan rates. High scan rates allow rapid
temperature gradient programs, resulting in shorter run times and increased sensitivity
(Gummer et al., 2009). Alternatively, the combination of two-dimensional GC with TOF-
MS has resulted in the development of very high resolution fast MS that can be used to
detect more metabolites than is possible using single queadrupole (Q), TOF and ion trap
mass analyzer.
One if the drawbacks of many GC-MS metabolomics analyses is the need to derivitise
metabolites before analysis. In contrast, many classes of polar metabolites can be analyzed
directly by LC-MS without derivitisation. LC systems interfaced with TOF mass analyzer
are now commonly used in metabolomics analyzes, delivering high throughput and high
10 Metabolomics

mass resolution analysis capability with mass accuracy approaching single digit ppm.
Recently developed instruments also allow rapid polarity switching between positive and
negative mode within a single run, reducing the need for multiple runs and cost per sample
(Gummer et al., 2009).

Analysis NMR MS
High throughput-metabolites No Medium
High throughput-samples; automation Yes No
Quantitative Yes Yes
Availability in clinic No No
Equipment cost High High
Maintenance cost Medium High
Per sample cost Low High
Required technical skills Yes Yes
Sensitivity Medium High
Reproducibility High Low
Data analysis automation Yes Yes
Identification of new metabolites Difficult Possible
Chemical exchange analysis Yes No
In vivo measueremnt Possible Impossible
Table 2. Comparison of characteristics of major experimental methods for metabolomic
analysis

LC-MS linear quadrupole, triple quadrupole (QQQ), QTrap and io trap mass analyzers have
also been utilized for global and targete metabolomics, but may be limited by mass accuracy
and mass resolution in identifying metabolites. However, the use of triple quadrupole and
QTrap mass analyzers in various selective ion scaning modes can be used to detect specific
metabolites or metabolite classes with high sensitivity and are particulary useful for targeted
metabolomic analysis.
CE-MS offers a complementary approach to LC-MS for analyzing anions, cations, and
neutral particles in a single run. Metabolites can be analyzed directly without derivitisation
and the chromatographic resolution and sensitivity of CE is very high. However, CE is less
frequently used for metabolomic analyzes tha LC-MS.
Vibrational spectroscopies are relative insensitive, but FTIR allows for high throughput
screening of biological samples in an unbiased fashion. Similar to NMR, water signals pose a
problem and must be subtracted electronically or attenuated total reflectance may be used.
Compared with the other methods it is one of the least sensitive, but its unbiasness to
compounds and ability to analyse large numbers of sample in a day makes it a plausible
method for screening purposes (Khoo and Al-Rubeai, 2007).
LC-MS-based instruments can be operated in direct infusion mode with no chromatographic
separation for measurement of the total mass spectrum for the mixture. The infusion can be
performed with either the LC autosampler or with and offline syringe pump. Ion trap, TOF,
Q-TOF, Orbitrap and FT-ICR-MS mass analyzers have been used with this mode of sample
delivery. This approach relies totally on the mass analyser to resolve isobaric metabolites
such as leucine or isoleucine. The key advantange of direct infusion analysis is the potential
Metabolomics and Mammalian Cell Culture 11

for automated high throughput sample analysis with both low and high mass resolution
mass analyzers.
Another beneficial experimental method for cell culture metabolomics analysis involves
stable isotope labeling followed by either MS or NMR measurement. This approach enables
pathway tracing, easier metabolite assignment and metabolic flux measurements. Isotopic
labeling has previously enabled detailed determination of pathways leading to the
production of specific metabolites and the development of the highly accurate mathematical
models of these pathways (Hollywood et al., 2006).

4. Aplications in mammalian cell culture – Study cases


4.1 Analysis of molecular mechanisms associated to the adaptation of NS0 myeloma
cell line to protein-free medium
The NS0 mouse myeloma cell line has become one of the most popular systems for large-
scale heterologous protein expression. For reasons of regulatory compliance, cost, batch
consistency, downstream processing, and material availability, industrial applications of
NS0 has moved towards serum or protein-free medium platforms (Barnes et al., 2000). For
serum- or protein-free cultivation, the cell culture medium is often supplemented with
lipids (derived from plant or synthetic sources) in addition to other protein supplements.
The effect of lipid supplementation on the physiology of hybridomas and myelomas has
been reported (Jenkins et al., 1992). NS0 cells are naturally cholesterol-dependent; not only is
their growth greatly facilitated by lipid supplementation, but is also dependent on provision
of cholesterol. NS0 cells capable of cholesterol-free growth can be isolated by selecting
mutant clones or by adaptation. Adaptation generally involves passaging cells over a time
period during which the serum concentration is decreased gradually (Sinacore et al., 2000).
Eventually, the resulting population develops the capability to grow in the absence of
serum. Different mechanisms underlying a cholesterol-dependent phenotype could include
the absence (or mutation) of a gene or a segment of gene along the cholesterol biosynthesis
pathway. There could be changes in the expression level of some proteins of the pathway
due to gene regulation or other control mechanisms. In addition to the specific gene
expression alterations along the cholesterol and lipid metabolism pathways, cholesterol
dependence could also be the result of insufficient precursor supply (Spens and Haggstrom,
2005).
The molecular mechanisms of host and recombinant NS0 cell lines that could be related to
the adaptation to protein-free medium are studied in this work. A quantitative study of
proteins with differential expression levels in four conditions (host NS0 cell line adapted
and non-adapted to protein-free medium, and a monoclonal antibody (Mab) transfectoma
producer NS0 adapted and non-adapted to the same protein-free medium) is reported. The
study is based on the use of the combination of two-dimensional electrophoresis and mass
spectrometry, and a novel quantitative proteomic approach, isobaric tagging for relative and
absolute quantification (iTRAQ). The metabolic study of these cell lines cultured in different
nutrient conditions is also reported. Taking into account the proteomic results and metabolic
analysis, a possible mechanism related to the adaptation of NS0 cell line to protein-free
medium is proposed.
12 Metabolomics

4.1.1 Results: Proteomic analysis


To characterize the changes associated with the adaptation to the protein-free medium 2DE
gels of protein extracts from the cell line cultured in PFHM II with or without 1% (v/v) FBS
were compared over a pI range of 3 to 10. Following adaptation to the protein-free medium,
78 spots changed their intensity by a factor of ≥2 from 1200 detected spots/ treatment.
Interestingly, the majority of differentially expressed proteins decreased their expression in
cells adapted to the protein-free medium. Fifty eight proteins were characterized by
MALDI-MS and/or LC-ESI-MS-MS. The identified proteins were grouped according to their
molecular function. Four major cellular pathways seem to be involved in the adaptation to
the protein-free medium: i) carbohydrate metabolism and energy production, especially
glycolysis and the Krebs cycle, ii) protein synthesis and folding; iii) membrane transport,
and iv) cell proliferation (de la Luz et al., 2007).
In order to increase the number of proteins related with cell cycle regulation, DNA
replication and lipids synthesis we used another strategy based in the isobaric labeling and
the subcellular fractionation. iTRAQ reagent technology is a newly developed method for
relative quantification of proteins from up to four samples. It has immense potential to
improve the sensitivity and quality of mass spectrometric analysis of the proteome. We were
able to identify and quantify 575 proteins simultaneously from the four states of culture.
Among these 575 proteins, 43% were identified by a single significant peptide per protein;
the rest were identified by at least two significant peptide of the same protein. The method
used in our case to analyze the differential expression levels between two or more
conditions was the locfdr. This method is a new approach to the problem of multiple
comparisons and controls the number of false positive differentially expressed proteins
below the user-specified threshold.
The standard deviation for each peptide value is obtained from the iTracker estimates, while
in the case of proteins the variant of t-statistic suggested by Efron was used (Jung et al.,
2006) to skip the inconvenience of those proteins that were identified by a single peptide
and have standard deviation zero. In all analysis the condition locfdr ≥0.2 was used to select
differentially expressed proteins. This threshold is a general accepted standard in fdr
applications; it means that to consider a protein as differentially expressed the
corresponding probability of being a false positive must be below 20%.
Following this approach we have found a set of 102 differentially expressed proteins. These
proteins were classified in different functions and locations according to the KEGG database
(de la Luz et al., 2008). According with the previous results four major cellular pathways
seem to be involved in the adaptation to the protein-free medium (Figure 2).

4.1.2 Results: Kinetic and metabolic analysis


The host and recombinant NS0 cell line were culture in serum-supplemented and protein-
free medium during 140 hours. Total cell number, viable cell number and viability were
determined. The specific growth rate is different between both cells, but there was a clear
decrease when both cell lines were cultured in absence of serum (Table 3). Intracellular
metabolite concentrations were calculated during exponential growth phase. In contrast
with previous reports, we found a lost of cholesterol auxotrophy in the host and
recombinant NS0 cell line adapted to PFHM. Other metabolites such as phospholipids and
Metabolomics and Mammalian Cell Culture 13

fructosamine involved in specific cellular processes like membrane biogenesis and


glycolipid metabolism changed their expression levels in adapted versus non-adapted cell
line. In order to check if the intracellular cholesterol concentrations increase in the adapted
cell line is a reversible process, cells were cultured in a medium supplemented with serum,
and the initial cholesterol levels were determined (de la Luz et al., 2008).

Fig. 2. Distribution of identified proteins taking in account their biological function

Table 3. Cellular growth parameters from host NS0 and recombinant NS0 cell lines adapted
and non-adapted to protein-free medium

Glycolysis is one of the most important metabolic pathways providing a source of


precursors and energy for the cell. Previous analysis by DNA microarray studies have
revealed a large number of genes involved in glycolysis, the pentose phosphate pathway
and the Krebs cycle to be down-regulated in host NS0 cell line cultured in the absence of
cholesterol (Seth et al., 2005). Ten proteins from glycolysis were found up-regulated in non
adapted NS0 cell line with respect to adapted. This result could indicate that the glycolysis
is a source of molecular precursors (cholesterol and phospholipids), especially in the
adapted cell line (Figure 3). The lactate production increase after the adaptation process
could be related with the higher lactate dehydrogenase enzyme activity (especific enzyme
activity NS0 adapted: 20.85 U*mL-1*cell-1, non-adapted: 12.31 U*mL-1*cell-1).
14 Metabolomics

Fig. 3. Metabolic model of the adaptation of NS0 cel line to protein free-medium obtained
from proteomic and metabolites analysis. Red: sub-expressed pathways. Blue: over-
expressed pathways.

With the aim to calculate the relative rate of glycolysis and glutaminolysis, intracellular
concentration of lactate and glucose were determined in the batch culture and the
relationships between lactate production and glucose consumption (qL/qG) were calculated
(figure 4). These results indicated that the lactate produced depend of the glycolysis and the
glutaminolysis. Taken into account protemic and metabolic results we have proposed a
metabolic mechanism where the glucose is used for the precursors synthesis. On the other
hand, the cell obtain the energy from glutamine degradation.
We used the flux balance analysis (FBA) in order to compare the results obtained with an
empiric metabolic network with the experimental results. In this study we used a reported
metabolic network (Ma and Zeng, 2003) with changes in the cholesterol reactions, where the
cholesterol synthesis pathways was eliminated in NS0 non-adapted. The comparison
between adapted and non adapted metabolic network showed changes in carbohydrate and
lipid metabolism, very similar with our previous experimental results. Also we analyzed the
metabolites that have influence in cellular growth when they are not present in the medium.
Glycine, tryptophan, phenylalanine, adenine, palmitic acid, glutamic acid, methinonine and
asparagine are relatefd with the increase of cellular biomass (data not shown).
Metabolomics and Mammalian Cell Culture 15

Fig. 4. Glucose consumption and lactate production during a batch culture of myeloma cell
line in presence and absence of serum. During the culture period, samples were taken
periodically for off-line analysis and media metabolites concentration were determined. The
relation between lactate production and glucose consumption is representative of the
cellular metabolic state, especially of the glycolysis efficiency

5. Conclusion
Data integration is not limited to flux data. Systems biology encompasses a holistic
approach to the study of biology and the objective is to simultaneously monitor all
biological processes operating as an integrated system. The use of the data obtained from
studies with different “omics” techniques is not simple. In addition, a single gene may code
for isoenzymes reacting with multiple metabolite substrates. The difficulty in determining
the timing of different events, that it, transcription and protein activity, also contribute to
the difficulty in integrating data. Hence in order for metabolomics to be used in systems
biology, novel strategies will need to be created. One step forward in such an integration
process is the functional assignments between protein/gene and metabolite within a system
of interest. This can be done by creating models where basic biochemical pathways are
modelled using static data (Khoo and Al-Rubeai, 2007). Second, time-dependent
concentrations of other types of components (transcriptomics and/or proteomics) will then
be incorporated followed by the reconstruction of the model with statistic data.
In contrast with previous results, changes in metabolic rates and biosynthetic machinery
with respect to the presence or not of serum in the culture medium were observed in this
study. The analysis was performed by two different ways. First, using iTRAQ reagents,
proteins with differential expression levels in two myeloma cell lines cultured in serum-
supplemented and serum-free medium were detected. These proteins belong to major
pathways related with glycolysis, protein synthesis and membrane transport. These
results are in accordance with previous results obtained using 2DE and the study of a
revertant
16 Metabolomics

NS0 cholesterol-independent (Seth et al., 2005). Second, the determination of consumption


and production of different metabolites like glucose, lactate, cholesterol, phospholipids and
phosphorous was performed. Differences in qL/qG were found between adapted and non-
adapted cell lines, similar to the results obtained by proteomics. A significant increase was
observed in the intracellular cholesterol concentration in the adapted cell lines. However,
when these cell lines adapted to PFHM were cultured in presence of serum, the intracellular
cholesterol levels decreased down to the initial conditions, indicating a possible epigenetic
mechanism.

6. References
Earle W.R. et al (1954) Certain factors limiting the size of the tissue culture and the
development of mass cultures. New York Academy of Sciences 58, 1000-1011
Lake-Ee Quek et al. (2010) Metabolic flux analysis in mammalian cell culture. Metabolic
Engineering 12, 161-171
Warburg O. (1956) On the origin of cancer cells. Science 123, 309-314
Fitzpatrick L. et al. (1993) Glucose and glutamine metabolism of a murine B-lymphocyte
hybridoma grown in batch culture. Applied Biochemistry and Biotechnology 43, 93-116
Jenkins H.A. et al. (1992) Characterization of glutamine metabolism in two related murine
hybridomas. Journal of Biotechnology 23, 167-182
Ljunggren J. and Haggstrom L. (1992) Glutamine limited fed-batch culture reduces the
overflow metabolism of amino acids in myeloma cells. Cytotechnology 8, 45-56
Ozturk S.S. and PalssonB.O. (1991) Growth, metabolic, and antibody production kinetics of
hybridoma cell culture: 1 Analysis of data from controlled batch reactors.
Biotechnology Progress 7, 471-480
Martinelle K. et al. (1998) Elevated glutamate dehydrogenase flux in glucose-deprived
hybridoma and myeloma cells: evidence from H-1/N-15 NMR. Biotechnology and
Bioengineering 60, 508-517
Korke R. et al. (2002) Genomic and proteomic perspectives in cell culture engineering.
Journal of Biotechnology 94, 73-92
Seow T.K. et al. (2001) Proteomic invetsigation of metabolic shift in mammalian cell culture.
Biotechnology Progress 17, 1137-1144
Seth G. et al. (2007) Molecular portrait of high productivity in recombinant NS0 cells.
Biotechnology and Bioengineering 97, 933-951
Smales C.M. et al. (2004) Comparative proteomic analysis of GS-NS0 murine myeloma cell
lines with variyng recombinant monoclonal antibody production rate. Biotechnology
and Bioengineering 88, 474-488
de la Luz K.R. et al. (2007) Proteomic analysis of the adaptation of the host NS0 myeloma
cell line to a protein-free medium. Biotecnología Aplicada 24, 215-223
de la Luz K.R. et al. (2008) Metabolic and proteomic study of NS0 myeloma cell line
following the adaptation to protein-free medium. Journal of Proteomics 71, 133-147
Oliver S.G. et al. (1998) Systematic functional analysis of the yeast genome Trends in
Biotechnology 16, 373–378
Beecher C.W. (2002) Metabolomics: A new "omics" technology. American Genomics -
Proteomics Technology
Metabolomics and Mammalian Cell Culture 17

Khoo S.H.G and Al-Rubeai M. (2007) Metabolomics as a complementary toolo in cell


culture. Biotechnolgy and Applied Biochemistry 47, 71-84
Hollywood K. et al. (2006) Metabolomics: current technologies and future trends. Proteomics
6, 4716-4723
Fiehn O. (2001) Metabolomics – the link between genotype and phenotype. Plant Mol. Biol.
48, 155-171
Viant M.R. et al. (2005) An NMR Metabolomic Investigation of Early Metabolic Disturbances
Following Traumatic Brain Injury in a Mammalian Model. NMR in Biomedicine 18,
507-516
Tweeddale H. et al (1998) Effect of slow growth on metabolism of Escherichia coli, as
revealed by global metabolite pool (‘‘metabolome’’) analysis. J Bacteriol 180, 5109–
5116
Buchholz A. et al. (2001) Quantification of intracellular metabolites in Escherichia coli K12
using liquid chromatographic electrospray ionization tandem mass spectrometric
techniques. Anal Biochem 295,129–137
Villas-Boas S.G. et al. (2005) Mass spectrometry in metabolome analysis. Mass Spectrometry
Reviews 24: 613 - 646.
Cuperlovic-Culf M. et al. (2010) Cell culture metabolomics: applications and future
directions. Drug Discovery Today 15, Numbers 15/16
Gerdtzen Z.P. et al. (2004) Non-linear Reduction for Kinetic Models of Metabolic Reaction
Networks. Metabolic Engineering 6, 140-154
Taoka S. and Banerjee R.(2002) Stopped-flow kinetic analysis of the reaction catalyzed by the
full-length yeast cystathionine beta-synthase. J Biol Chem. 277(25), 22421-22425
ap Rees T. and Hill S.A. (1994) Metabolic control analysis of plant metabolism. Plant, Cell &
Environment 17, 587-599
Fienh O. et al. (2000) Identification of uncommon plant metabolites based on calculation of
elemental compositions using gas chromatography and quadrupole mass
spectrometry. Anal Chem. 72, 3573–3580
Orth H.C. et al. (1999) Isolation, purity analysis and stability of hyperforin as a standard
material from Hypericum perforatum L. J Pharm Pharmacol 51, 193-200
Dietmair S. et al. (2010) Towards quantitative metabolomics of mammalian cells:
development of a metabolite extraction protocol Anal Biochem. 404, 155-64
Dunn W.B. and Ellis D.I. (2005) Metabolomics and systems biology: making sense of the
soup. Curr. Opin. Microbiol. 7, 296–307
van Greef J. et al. (2004) The role of analytical sciences in medical systems biology. Curr Opin
Chem Biol 5, 559–565
Lindon J.C. et al. (2007) The handbook of metabonomics and metabolomics Elsevier
Koek M.M. et al. (2010) Semi-automated non-target processing in GC × GC–MS
metabolomics analysis: applicability for biomedical studies Metabolomics 7, 1–14.
Ala-Korpela M. (2008) Critical evaluation of H NMR metabonomics of serum as a
methodology for disease risk assessment and diagnostics Clinical Chemistry and
Laboratory Medicine 46, 27–42.
Detmer K. et al. (2007) Mass spectrometry-based metabolomics Mass Spectrometry Reviews 26,
51-78
18 Metabolomics

Griffin J.L. (2003) Metabonomics: NMR spectroscopy and pattern recognition analysis of
body fluids and tissues for characterization of xenobiotic toxicity and disease
diagnosis Curr. Opin. Chem. Biol. 7, 648-654
Barnes L. Et al. (2000) Advances in animal cell recombinant protein production: GS-NS0
expression system. Cytotechnology 32, 109-123
Jenkins H. et al. (1992) Characterisation of glutamine metabolism in two related murine
hybridomas Journal of Biotechnology 23, 167-182
Gummer J. et al. (2009) Use of mass spectrometry for metabolite profiling and metabolomics
Australian Biochemist 40, 5-8
Sinacore M. et al. (2000) Adaptation of mammalain cells to growth in serum-free media
Molecular Biotechnology 15, 249-257
Spens E. and Haggstrom L. (2005) Defined protein and animal component-free NS0 fed-
batch culture Biotechnology and Bioengineering 98, 6
Jung Y. et al. (2006) Identifying differentially expressed genes in meta-analysis via Bayesian
model-based clustering Biomolecules Journal 48, 435-450
Seth G. et al. (2005) Large-scale gene expression analysis of cholesterol dependence in NS0
cells Biotechnology and Bioengineering 90, 552-567
Ma H. and Zeng A. (2003) Reconstruction of metabolic networks from genome data and
analysis of their global structure for various organisms Bioinformatics 19, 270-277
2

Quantitative Metabolomics and Its Application


in Metabolic Engineering of Microbial Cell
Factories Exemplified by the Baker’s Yeast
Mario Klimacek
Institute of Biotechnology and Biochemical Engineering, Graz University of Technology
Austria

1. Introduction
The baker’s yeast Saccharomyces cerevisiae and its beneficial properties have been recognized
very early by human beings. It has been used in the making of alcoholic beverages, bread
and cake long before the term biotechnology has been coined. In addition to its great
importance in food industry S. cerevisiae strains are nowadays applied in many other fields
for example in the production of bio-fuels from corn or sugar containing crops, in the bio-
sorption of heavy-metals from sewage, in pharmaceuticals or in the production of precursor
compounds for the synthesis of pharmaceuticals or fine chemicals. As a consequence S.
cerevisiae developed to one of the most important and best investigated microbial cell
factories for the industrial (white) biotechnology. Furthermore S. cerevisiae is an important
model organism used to elucidate the underlying molecular mechanistic principles that are
involved in complex diseases (cancer or diabetes) and metabolic disorders (Castrillo and
Oliver 2005; Castrillo and Oliver 2006; Nielsen and Jewett 2008). Other important features of
S. cerevisiae that led to its multifaceted applicability in industry and R&D constitute its
GRAS (generally recognized as safe) status and that cells are very easy to cultivate and are
readily available.
The physiology of S. cerevisiae under various environmental conditions has been
investigated intensively in the last 140 years (Racker 1974). The baker’s yeast exhibits
some very interesting physiological features that render it unique among all other
microorganisms. It grows nearly equally fast under aerobic and anaerobic conditions with
glucose as the sole carbon source (Nissen et al. 2000a; Visser et al. 1990). Under aerobic
conditions and at glucose concentrations above 100 mg/L biomass formation is
accompanied by the production of ethanol as a consequence of an overflow metabolism at
the pyruvate node (Crabtree-effect, (Crabtree 1928)). After depletion of glucose the
ethanol initially formed by the overflow metabolism is further converted into biomass
under aerobic conditions (Diauxie). Under anaerobic conditions about 90% of glucose
carbon is converted into ethanol and CO2. The rate of glucose utilization and the specific
ethanol yield is higher under anaerobic conditions as compared to the sugar conversion
rate and ethanol yield under aerobic conditions (Pasteur-effect, (Racker 1974)). It can
reduce a number of keto-compounds to the corresponding chiral alcohols that represent
20 Metabolomics

interesting precursors for pharmaceuticals (Csuk 1991). It can grow as a diploid as well as
a haploid which highly facilitates genetic manipulation and permits high-throughput
genetic engineering.
Considering the enormous early interest in studying and understanding the physiology of
S. cerevisiae long before modern omics techniques have been developed, it is not very
surprising that it was the baker’s yeasts genome that was the first within the domain of
eukaryotes that was completely sequenced. Genomic and biological information about S.
cerevisiae molecular biology is comprehensively collected at the Saccharomyces Genome
Database (SGD, http://www.yeastgenome.org/). Driven by the knowledge of the
complete genomic sequence and by the steadily increasing availability of tools developed
for genetic engineering, S. cerevisiae became a key work horse and the representative
eukaryotic model organism in every modern discipline within the biosciences such as
molecular and cell biology, functional genomics, systems biology or metabolic and
synthetic engineering. Today’s genetic work with S. cerevisiae cells is highly alleviated by
the presence of a wide spectrum of established yeast molecular biology tool kits and
availability of many wild-type and mutant strain (e.g. knock-out strains) collections as
well as plasmid collections containing S. cerevisiae ORFs, gene deletion markers or
promoter sets and many more, offered by commercial sources such as EUROSCARF
(http://web.uni-frankfurt.de/fb15/mikro/euroscarf/index.html), Open biosystems
(http://www.openbiosystems.com/Products/) or Addgene (http://www. addgene.
org/).
The commercial establishment of genetic manipulation techniques paved the way for S.
cerevisiae to be exploited in the field of metabolic engineering. Various novel recombinant
designer strains capable of either selective formation of one desired product or of producing
heterologous compounds or endogenous products from new resources (waste or renewable
materials) emerged in the last decades. Metabolic engineering efforts based on S. cerevisiae
are comprehensively summarized elsewhere and the interested reader is referred to (Bettiga
et al. 2010; Nevoigt 2008). A collection of engineered substrate utilization and heterologous
or homologous product formation pathways is given in Table 1.
The corresponding underlying engineering principles can be basically broken down into 4
strategies as depicted in Figure 1 panel A-D. Elucidation of the appropriate engineering
approach represents the most important step in designing novel cellular properties and
targets on the identification of reaction(s) or even entire pathways that are suited for the
anticipated metabolic engineering objective. Relevant reaction(s) and associated gene(s) can
be extracted by thorough screenings of literature data (US National Library of Medicine
(http://www.ncbi.nlm.nih.gov/pubmed), SciFinder (https://scifinder.cas.org/) or Web of
Knowledge (http://wokinfo.com/)) and online databases (KEGG the Kyoto Encyclopedia
of Genes and Genomes (http://www. genome.jp/kegg/), the enzyme database BRENDA
(http://www.brenda-enzymes.org/), or the SIB bioinformatics Resource Portal ExPASy
(http://www.expasy.ch/).
To increase the probability of engineering success identified targets can be subjected to in
silico modeling by employing mathematical models like restricted flux balance analysis
(FBA) based on a genome-scale stoichiometric network to verify their compatibility with the
underlying metabolic network (Cvijovic et al. 2010; Selvarajoo et al. 2010).
Quantitative Metabolomics and Its Application in
Metabolic Engineering of Microbial Cell Factories Exemplified by the Baker’s Yeast 21

Substrate targets Applications References


(Jeffries and Jin 2004;
Xylose bio-ethanol Petschacher and Nidetzky
2008; van Maris et al. 2007)
L-Arabinose bio-ethanol (Wisselink et al. 2007)
Lactose / whey bio-ethanol (Domingues et al. 2010)
Galactose bio-ethanol (Lee et al. 2010)

Product targets

Insulin and insulin


pharmaceuticals (Kjeldsen 2000)
precursors
Hepatitis B antigen pharmaceuticals (Kuroda et al. 1993)
Cyanophycin biopolymer (Steinle et al. 2009)
(Abbott et al. 2009; Ishida et
Organic acids chemical building blocks
al. 2006; Raab et al. 2010)
n-Butanol bio-fuel (Steen et al. 2008)
(Asadollahi et al. 2010;
Sesquiterpenes pharmaceuticals and food
Jackson et al. 2003)
Carotenoids pharmaceuticals and food (Ukibe et al. 2009)
Diterpenoids pharmaceutical industry (Dejong et al. 2006)
Triterpenes pharmaceutical industry (Madsen et al. 2011)
Polyketides pharmaceutical industry (Mutka et al. 2006)
Five-carbon sugars /
food ingredients (Toivari et al. 2007)
alcohols
Ethylene synthetic polymers (Pirkov et al. 2008)
Flavonoids, stilbenoids pharmaceuticals and food (Trantas et al. 2009)
SO2 beer flavor stability (Yoshida et al. 2008)
Ethanol bio-fuel (Nissen et al. 2000b)
Table 1. Collection of metabolic engineering targets in S. cerevisiae

To unravel reaction(s) or pathways instead that would compromise substrate conversion


and/or production of a desired product in silico modeling by for example FBA (Bro et al.
2006) or MOMA (minimization of metabolic adjustment) (Asadollahi et al. 2009) have been
applied and produced potential candidate reactions which otherwise would have been often
overlooked (Cvijovic et al. 2010). The open-source software platform OptFlux developed
especially for in silico driven metabolic engineering is available at http://www.optflux.org/
(Rocha et al.).
Metabolic integration of a novel pathway is selected when the utilization of a new substrate
(Fig. 1 A) or synthesis of a new product is intended (Fig. 1 B). New in this context means
that the original, not engineered, cells are genetically not programmed to perform these
reactions. Formation of an undesired side-product (Fig. 1 C) can be attributed to a split in
carbon flux at the related node (C) into a productive (CP) and undesired flux (CD).
Reasons for such flux partition compromising efficient production of the product (P) can be
often traced back to the presence of an enzyme or differently regulated isoenzyme
competing for the same substrate S or a promiscuous enzyme that in addition to the desired
substrate is also active with other substrates (Fig. 1 D).
22 Metabolomics

Fig. 1. Typical metabolic engineering principles based on rational design (panels A – D) are
linked to a suggested experimental work-flow to unravel limiting metabolic sites. Panels A-
D refer to enabling of substrate utilization (A) or product formation (B), preventing side
product formation by deletion and/or overexpression of an endogenous enzyme (C),
increasing selectivity of a substrate promiscuous enzyme (D); Substrate A, intermediate B,
product P and enzymes E new to the network are indicated in grey. Overexpression of an
endogenous enzyme is depicted by a grey e. Knock outs are indicated by grey x’s. Subscripts
of rate constants v given as numbers and small letters refer to fluxes based on stoichiometry
(solid arrows) and individual reaction rates of enzymes (dotted arrows), respectively.

Directing the carbon flux towards P can be afforded by deletion of respective gene(s),
overexpression of enzymes participating in the productive branch, or replacing the
corresponding activity by a less regulated or more selective one. Furthermore unbalanced
carbon usage between reaction partners participating in this pathway and/or in the
recycling of, for example cofactors, can result in accumulation and release of a pathway
intermediate (Krahulec et al. 2009; Krahulec et al. 2010). In this case fine-tuning of all
activities involved based on for example a metabolic control analysis (MCA) or kinetic
modeling analysis is required to minimize or even completely prevent by-product
accumulation (Parachin et al. 2011).
Aside from rational design stochastic methods based on inverse metabolic engineering have
been developed for S. cerevisiae to identify key target reactions and associated gene
sequences enabling the desired new cellular property (Bailey et al. 2002; Bengtsson et al.
2008; Bro et al. 2005; Hong et al. 2010; Jin et al. 2005; Lee et al. 2010). Differently, methods
targeting on the induction of a cellular property, such as growth, increase of substrate
conversion rate or enhancing resistance to environmental stress, that is hardly to capture by
in silico design because of its highly intricate metabolic relations that have to be satisfied,
rely on the cellular adaptability to a certain environmental stress by evolution (Cakar et al.
Quantitative Metabolomics and Its Application in
Metabolic Engineering of Microbial Cell Factories Exemplified by the Baker’s Yeast 23

2009; Cakar et al. 2005; Garcia Sanchez et al. 2010; Kuyper et al. 2005; Sonderegger and Sauer
2003; Wisselink et al. 2009).
In the course of establishing systems biology various high-throughput omics techniques such
as transcriptomics, proteomics, fluxomics and others have been developed with the objective
to comprehensively analyze cellular physiology at all molecular levels (DNA, RNA, protein,
flux, and metabolite). Data-driven analysis is often exploited to unravel novel interrelations at
the various molecular levels or to obtain a more insightful (quantitative) understanding of
cellular processes. It is obvious that metabolic engineering can greatly benefit from the
integration of omics techniques in the design of improved microbial cell factories (Nielsen and
Jewett 2008). The various omics tools have helped to increase understanding about how cells
regulate, communicate and adapt to different environmental conditions.
Depending on the metabolic engineering objective the appropriate omics tool or a
combination should be selected after due consideration. For example transcriptome analysis
provides a holistic image of mRNA molecule pattern and levels but do not tell us anything
about metabolic fluxes. Optimization of the flux towards a specific metabolite however
represents one of the major goals in metabolic engineering. Metabolic flux analyses based on
stoichiometric models or 13C-isotopomer analysis (provided that cells can grow under the
environmental conditions used) are useful tools in this respect (Nielsen and Jewett 2008). To
understand the underlying mechanistic relationships between the flux through a particular
pathway and the enzymes forming the pathway, providing the relevant information for
strain design, detailed knowledge about enzyme-metabolite interactions are required.
Consequently quantitative information about metabolites involved together with detailed
knowledge of kinetic properties of participating enzymes is mandatory. Within the omics
family metabolomics represents the youngest member. This is basically due to the facts that
metabolites vary greatly in their physico-chemical properties (polarity, acidity, reactivity,
and stability) and are present in a large dynamic concentration range which make it almost
impossible to record the entire metabolome on a single analytical platform. Another
challenge represents the generation of reliable and representative metabolite data from
biological samples. Cell-wall leakage, instabilities and losses of metabolites throughout the
sample work-up, or strong matrix effects in the MS analysis are a few of the many causes
impairing metabolite data and as a consequence adulterate molecular mechanistic
interpretation. Nevertheless in the last years much progress has been made due to
enormous efforts of the yeast research community to overcome these obstacles. Protocols of
unbiased sample-work-up and different analytical platforms are available today that can
cover more than 100 compounds quantitatively.
This review presents current accepted protocols and techniques that enable acquisition of
absolute quantitative metabolite data from S. cerevisiae cells. The second part focuses on how
quantitative metabolite data can help in the development of improved microbial cell factories.
However, before going into the details some definitions of terms used in metabolome
analysis should be reminded (Nielsen 2007). Metabolite profiling targets on the qualitative
or semi-quantitative analysis of specified metabolites or groups of metabolites. In contrast in
metabolite target analysis selected metabolites are quantified. If the entire metabolome or a
fraction of it is addressed (or as many metabolites as possible) qualitatively or quantitatively
we speak of metabolomics or quantitative metabolomics.
24 Metabolomics

2. Data acquisition for quantitative metabolomics in Saccharomyces


cerevisiae
Determination of unbiased intracellular concentrations of metabolites is without doubt a
prerequisite for serious interpretation of cellular properties at the molecular – kinetic level.
Unbiased refers here to the physiological state, the sample work-up and preparation for
metabolite analysis and metabolite quantification and calculation of intracellular molar
concentration. Consequently four experimental tasks that have to be fulfilled can be
formulated as follows:
 Harvesting and quenching of cells at a predefined physiological state (representative)
and separation from extracellular compounds (exometabolome) without leakage of
intracellular metabolites (quantitative)
 Destruction of metabolic activity and complete extraction of metabolites by maintaining
the metabolite composition quantitatively unchanged, concentration and if required
chemical preparation for metabolite analysis
 High-throughput quantitative analysis by employing the appropriate instrument
 Relation of molar concentrations obtained from quantitative analysis to cell specific
parameters such as cell dry weight (µmol / gCDW) or cell number (µmol / cell) or if
absolute molar intracellular concentrations are required to the cell (compartment)
volume.
However, before we go into the details and hurdles of each task it should be emphasized
here that altering the cellular network by metabolic engineering will always result in a quasi
new strain with completely new properties and behaviors. In the worst case the complete
sample work-up protocol established for the wild-type strain might not be applicable for the
mutant strains. It is therefore strongly recommended that protocols already established for
the absolute quantification of metabolites have to be re-assessed and verified for the created
strains.

2.1 Representative harvesting and quantitative quenching


The concentration of a metabolite in the cell is not directly linked to genes but is determined by
its formation and utilization rates. Conversion rates in turn depend on the enzymes associated
catalytically with this metabolite and their kinetic parameters with respect to this metabolite as
well as effectors (inhibitors, activators). In particular metabolites from catabolic reactions and
reactions involved in energy metabolism display high turnover rates. For example for the
frequently used metabolite ATP (~16% of all reactions present in a genome-scale
stoichiometric model of S. cerevisiae involve ATP (Förster et al. 2003)) turnover rates of ~1.5
mM/s were reported (Rizzi et al. 1997). Hence, quenching of metabolic activity within a very
short time window is required without altering the metabolomic state of the sample. Another
highly desirable feature is that the extracellular environment containing substrates, products,
salts and the exometabolome, that can affect subsequent metabolite analysis, is separable from
the cells without losing any intracellular metabolites due to disruption of or leakage through
the cell wall. Different to for example E. coli the cell wall of S. cerevisiae is less leaky and both
requirements can be achieved for S. cerevisiae cells by spraying a defined volume of cell
suspension into an appropriate quenching solution at sufficiently low temperatures. It was
Quantitative Metabolomics and Its Application in
Metabolic Engineering of Microbial Cell Factories Exemplified by the Baker’s Yeast 25

found that the volume ratio of sample to quenching solution affects the quenching quality and
a ratio of at least 1 to 5 was suggested (Canelas et al. 2008a).
Today two methods cold methanol and cold glycerol-saline have achieved wide
acceptance within the metabolomics community (Canelas et al. 2008a; Villas-Bôas and
Bruheim 2007). See Fig. 2 for details. In addition to the quenching temperature and time
or the volume ratio between sample and quenching solution, the time between
quenching and separation by centrifugation and the centrifugation time can influence
metabolic activity and metabolite leakage significantly. Harvesting by rapid sampling is
often coupled to quenching. Rapid sampling is especially important in continuous
steady-state cultivations and for pulse-experiments in which changes of intracellular
metabolite pools induced by a certain environmental impulse are analyzed at the sub-
second time scale. Ingenious devices have been developed in the last years that enable
rapid sampling and quenching simultaneously at the millisecond scale. The various
manual, semiautomatic and fully automatic rapid sampling techniques and their pros
and cons have been comprehensively discussed and the reader is referred to (Reuss et al.
2007; van Gulik 2010; Villas-Bôas 2007a). A disadvantage in this context is that most of
these devices are not available on the market and therefore not accessible to the scientific
community. Commercial accessibility however would be of great importance in the
context of comparability, reproducibility and standardization in quantitative
metabolomics studies. In batch cultivations manual transfer of the cell suspension to the
quenching solution by using a pipette or a syringe is widely accepted as environmental
conditions might not change significantly during the sample transfer (3 – 6 s) (Villas-
Bôas 2007b). These assumptions might hold for anaerobic or microaerobic conditions but
should be reconsidered in case of aerobic cultivations for which for example the O2/CO2
ratio may vary and induces changes (oxygen limitation) in cell metabolism during
sampling.

2.2 Quantitative metabolite extraction


The next challenge represents the quantitative extraction of all metabolites or at least of
those to be of interest without reactivating any metabolic activity, enabling chemical
reactions and minimizing metabolite disruption. Metabolic activity and chemical reactions
can be controlled by the temperature. The addition of a denaturing (inactivating) agent
bears some risks as it might be also reactive with metabolites or provides the environment
(pH) for chemical reactions (Villas-Bôas 2007b). Control of metabolite degeneration or
reactivity during extraction and in the following process steps represents a complex and
difficult task. Furthermore metabolite degeneration is a highly specific process and
strongly depends on the metabolite species, extraction solution and extraction parameters
(pH, T). For example the redox cofactors NAD+ and NADP+ are stable at acid to neutral
pH even at high temperatures (80-90°C) for 3 minutes (Klimacek et al. 2010). On the
contrary their reduced counterparts are highly unstable under acidic conditions with
NADPH generally less stable than NADH (Chaykin 1967; Lowry et al. 1961). At high pH
where NAD(P)H are stable NAD(P) in turn destruct rapidly (Lowry et al. 1961).
Decomposition can also be catalyzed by phosphate a prominent compound in metabolite
extracts (van Eunen et al. 2010) by forming an adduct with NADH across the pyridine
group (Alivisatos et al. 1964; Chaykin 1967).
26 Metabolomics

Fig. 2. Collection of state-of-the-art protocols for leakage-free quenching and quantitative


extraction. Relevant literature and details can be found in the text.

It is obvious from this example considering just four metabolites that the ideal extraction
procedure with which the complete metabolite consortium is extractable without any losses
may not exist and calls for a compromise in the selection of conditions used for metabolite
extraction. This “inconvenience” however can be circumvented, provided that subsequent
metabolite detection is based on mass spectroscopy, by the addition of an aliquot of U-13C-
labled internal standard (IS) compounds to the biomass subsequent to quenching or prior to
metabolite extraction (Büscher et al. 2009; Canelas et al. 2009; Klimacek et al. 2010; Mashego
et al. 2004; Wu et al. 2005). Metabolite losses due to incomplete quantitative work-up of
samples can be addressed by application of one selected IS compound. However metabolite
specific instabilities, matrix effects, ion suppression, non-linear responses and day-to-day
variations can be only identifed and appropriately corrected for by the addition of U-13C-
labled IS. A representative mixture of labeled metabolites can be easily prepared from S.
cerevisiae wild-type and/or mutant cells cultivated in standard mineral medium
supplemented with U-13C-labled substrate (glucose, fructose, galactose,…) under the
cultivation condition selected and by respective appropriate quenching and extraction
procedures. Internal referencing by using an IS displaying a metabolite composition that is
representative for the cellular state to be studied should be always taken into account
because intracellular metabolite levels can vary considerably in dependence on the
cultivation conditions used or on the cellular alterations introduced by pathway engineering
(Klimacek et al. 2010).
Quantitative Metabolomics and Its Application in
Metabolic Engineering of Microbial Cell Factories Exemplified by the Baker’s Yeast 27

Various extraction protocols with respect to extraction solvent (acids, bases, ethanol or
methanol, organic solvents), buffered or non-buffered solutions, pH, temperature, etc have
been tested (Villas-Bôas 2007b), evaluated and verified for S. cerevisiae cells in terms of
metabolite coverage, efficacy and recovery (Klimacek et al. 2010) in the last years (Canelas et
al. 2009; Villas-Bôas et al. 2005b). Today three extraction procedures have achieved some
acceptance and are likewise used within the yeast research community (see Fig. 2 for
details). That is boiling ethanol (BE; pioneered for S. cerevisiae cells by (Gonzalez et al. 1997)),
chloroform-methanol (CM; pioneered for S. cerevisiae cells by (de Koning and van Dam
1992)) and to some extent freeze-thawing in methanol (FTM; pioneered for S. cerevisiae cells
by (Villas-Bôas et al. 2005b) for which however controversial results with respect to its
applicability are present in the literature. While (Villas-Bôas et al. 2005b) found extraction
performance of FTM sufficient, others (Canelas et al. 2009) concluded that FTM cannot
effectively prevent metabolite conversion throughout the extraction process and considered
FTM therefore as not appropriate for metabolite extraction. Differences in evaluation criteria
and growth conditions were used as a basis to explain the different outcomes. It should be
however noted that (Canelas et al. 2009) investigated metabolite extraction performances
from S. cerevisiae cells grown under two different physiological conditions (glucose
limitation and glucose saturation; a bioreactor coupled to a rapid sampling device was
used), used identical U-13C-labled compounds as IS and three different analytical methods
for quantification of a broad range of different compounds. Quality and metabolite recovery
of FTM instead was judged by (Villas-Bôas et al. 2005b) by the application of an IS mixture
composed of compounds each a representative for a substance class analyzed. The mixture
was added to the quenched cells prior to extraction. Cells were cultivated under aerobic
conditions in shake flasks and metabolites were quantified by established GC-MS after
metabolite derivatization with methyl chloroformate. In Fig. 2 brief descriptions of
respective protocols are compiled. A broad spectrum of compounds covering a wide range
of different chemical properties such as acidity, polarity, size and responsiveness can be
addressed with either of these extraction protocols. Details with respect to extraction
method specific component coverage can be extracted from (Buescher et al. 2010; Büscher et
al. 2009; Canelas et al. 2008a; Canelas et al. 2009; Klimacek et al. 2010; Villas-Bôas and
Bruheim 2007; Villas-Bôas et al. 2005b).

2.3 Quantification of intracellular metabolites


Approximately 600 metabolites are present in S. cerevisiae cells and their concentrations
range from sub-µM to mM (Nielsen 2007). Their levels vary considerably in dependence on
the environmental conditions applied or mutation introduced. Hence analysis tools suited
for the determination of intracellular metabolites should be able to cover quantitatively as
many metabolites as possible in wide concentration ranges.
Early studies that have focused on analyzing intracellular metabolites used enzymatic
assays (Ciriacy and Breitenbach 1979; de Koning and van Dam 1992; Gonzalez et al. 1997;
Grosz and Stephanopoulos 1990; Kötter and Ciriacy 1993; Theobald et al. 1997) or NMR
analysis (den Hollander et al. 1981; Navon et al. 1979; Shanks and Bailey 1988) for
quantification of a few compounds present in the cell. To increase the coverage towards a
holistic quantitative record of the endometabolome enormous efforts were put on the
development of new methods and techniques in the last years. In particular mass
28 Metabolomics

spectrometry revealed to be excellently suited in this respect and analytics of metabolite


targeting switched therefore from an enzyme assay- or NMR-based to a mass spectrometry-
based technique. Today three platforms dominate the analytical part of metabolomics. That
is mass spectrometric detection in the form of a single, tandem or triple quadrupole or
orbitrap mass spectrometer (MS) coupled via an electrospray ionization source (ESI) to a
component separation device such as gas chromatography (GC), reverse-phased ion-pairing
or anion exchange liquid chromatography (LC) or capillary electrophoresis (CE). Cross-
platform comparison with respect to quantitative metabolomics revealed LC as the best
suited separation technique for analysis on a single platform in terms of versatility and
robustness. It was suggested by the authors that it is best complemented by the use of the
GC platform (Büscher et al. 2009). More than 100 metabolites could be successfully
quantified by GC-MS (~100 metabolites, (Villas-Bôas and Bruheim 2007; Villas-Bôas et al.
2003; Villas-Bôas et al. 2005b; Villas-Bôas et al. 2005c)), reverse phase ion pairing LC coupled
to a triple quadrupole (138 metabolites, (Buescher et al. 2010)) or an orbitrab mass spec (137
metabolites, (Lu et al. 2010)). Especially the LC-MS platform has been investigated
intensively with respect to quantitative metabolite coverage, sensitivity and robustness and
revealed to be very suited for the comprehensive analysis of the central carbon metabolism
(Buescher et al. 2010; Büscher et al. 2009; Lu et al. 2010). Almost all metabolites involved in
glycolysis, pentose phosphate pathway and TCA cycle could be addressed. In addition
amino acids and their precursors, redox cofactors, nucleotides, coenzyme A esters and many
more can be analyzed in one sample run in approximately half an hour (Buescher et al. 2010;
Lu et al. 2010). Current LC-techniques used in metabolomics are however limited to water
soluble analytes (Buescher et al. 2010).
As mentioned above absolute quantification of intracellular metabolites is indispensably
linked to the use of U-13C-labled internal metabolite standards. Consequently the number
of compounds to be analyzed doubles which makes data analysis more demanding.
Residual amounts of substrates, products and ionic components in the labeled and
unlabeled metabolite extracts can significantly alter the elution profile and ionization
characteristics of compounds analyzed (Buescher et al. 2010). These so called matrix
effects are hardly to predict and are typically experienced as an increase of the base line
signal associated with a high signal-to-noise ratio and as shifts in metabolite-specific
retention times. Consequently peak-to-peak resolution and base line separation of peaks
can become badly affected. Exacerbate and tedious manual peak integration is in these
cases required. Alternatively complex often erroneous peak deconvolution algorithms are
applied. To reduce matrix effects to a minimum demands preparation of “clean” -
meaning free of disturbing media compounds - metabolite extracts for both biological
samples and IS.
The recommended procedure for internal referencing by identical U-13C-labled compounds
involves addition of a defined volume of IS to the biological sample (prior to extraction) and
to all analytical standard dilutions (minimum 6 dilutions). A standard mixture containing all
the compounds at known concentrations to be analyzed is commonly used. As some
metabolites are not very stable it is recommended to store a master mixture containing just
the stable compounds and add the sensitive components prior to analysis. Metabolites are
quantified by comparing the ratio of 12C- to 13C- signals with the 12C/13C signals of the
representative standard compounds.
Quantitative Metabolomics and Its Application in
Metabolic Engineering of Microbial Cell Factories Exemplified by the Baker’s Yeast 29

2.4 Calculation of molar intracellular metabolite concentrations


The ultimate goal of quantitative metabolomics is the presentation of intracellular
metabolite pools in the form of absolute molar concentrations. Only the knowledge of molar
concentrations enables reliable integration of metabolome data in thermodynamic analysis,
application in MCA based on fundamental enzyme kinetics or kinetic modeling. In the last
step of absolute quantification of intracellular metabolite pools, the unbiased molar
concentrations of metabolites obtained by the methods described above have to be therefore
somehow related to the cell volume. For this reason metabolite concentrations and cell
volume are based on the cell dry weight (CDW) producing specific parameters for
metabolite concentrations in µmol/gCDW (= [metabolite]*volume of metabolite extract /
([dry cells]*volume of biological sample)) and for the cell volume in mL/gCDW. By
dividing these parameters the intracellular metabolite concentration in mM is eventually
obtained. Hence accurate determination of the CDW and cell volume is mandatory for
calculation of reliable intracellular metabolite data and for molecular mechanistic
interpretation relying on this data. Different methods can be found in the literature. Briefly,
first the physiological state has to be specified at which the CDW should be analyzed.
Second an aliquot of cell suspension is separated from the medium by vacuum filtration or
centrifugation. The cell pellet is washed with ice-cold water or physiological NaCl solution
to remove residual medium components and subsequently dried at 100-105°C until
constancy of mass is verified. One should however keep in mind that application of NaCl
can compromise resulting CDW values significantly when low amounts of biomass are
addressed. The volume of S. cerevisiae cells at a particular physiological state can be
determined by applying a Coulter counter analyzer or by microscopic techniques (Lord and
Wheals 1981; Tamaki et al. 2005).
The cell volume of the baker’s yeast was found to depend on the growth conditions and
environmental parameter settings and can vary considerably. For example, the cell volume
varies by a factor of 2-3 (16 – 42 µm3) with doubling time (Lorincz and Carter 1979; Tyson et
al. 1979) and the type of substrate metabolized (Johnston 1977; Tamaki et al. 2005). The
lower the doubling time the larger the cells (Tyson et al. 1979). Cells grown on glucose, the
most favored carbon source, are larger than those grown on a nonfermentable carbon source
(Johnston et al. 1979; Lorincz and Carter 1979; Porro et al. 2003; Tyson et al. 1979). Cells
cultured in the presence of ethanol showed an enlarged size (Kubota et al. 2004). In contrast
nitrogen starved cells are abnormally small (Johnston 1977). The level of
repression/derepression also contribute significantly to the cell size (Mountain and Sudbery
1990). Interestingly therefore that on the basis of the more relevant parameter for
quantification the specific cell volume, S. cerevisiae cells do not show significant variation
with the growth rate (Brauer et al. 2008) and values in the range of 1.5 – 1.9 mL/gCDW can
be found in the literature for the strain CEN.PK 113-7D (Canelas et al. 2011; Cipollina et al.
2008; van Eunen et al. 2009). A slightly higher value of 2.38 mL/gCDW has been reported
for another strain CBS 7336 (ATCC 32167) (Ditzelmüller et al. 1983). We can conclude that
although the cell volume is highly sensitive to conditions applied the specific cell volume is
rather constant. Nevertheless a 1.6-fold higher specific cell volume results in likewise lower
metabolite concentrations which in some cases (e.g. [substrate] < Km) may have an influence
on the data interpretation. In the light of metabolic engineering it is hard to predict whether
this “constant” likewise translates to recombinant cells. For example S. cerevisiae cells
30 Metabolomics

adapted to high ethanol concentrations displayed an altered cell size (Dinh et al. 2008). Or
overexpressing mannitol-1-phosphate dehydrogenase (M1PDH) in S. cerevisiae to produce
mannitol from glucose caused a substantial increasing of the size of cells (Costenoble et al.
2003).
As for all eukaryotic organisms S. cerevisiae metabolism is compartmented (cytosol,
mitochondrion, vacuoles) which poses a problem for the accurate determination of
concentrations of relevant intracellular metabolites. Current techniques for extracting
metabolites and isolating organelles do not allow for absolute separation from the cytosol
without altering the respective metabolite composition and pattern. Indirect strategies based
on metabolic engineering or on fundamental thermodynamic principles have been
developed to address this obstacle and gave first preliminary and semi-quantitative insights
into the distribution of metabolites between cytosol and mitochondrion.
Functional expression of M1PDH from E. coli in S. cerevisiae was used as indicator reaction to
determine the cytosolic free NAD to NADH ratio (Canelas et al. 2008b). M1PDH catalyzes the
reversible NAD(H)-dependent interconversion of fructose 6-P (F6P) and mannitol 1-P (M1P).
This reaction is directly connected to the central carbon metabolism and represents a dead-end
reaction in the metabolism of yeast under the conditions applied in this study. Based on the
assumption that the M1PDH reaction is at equilibrium the authors were capable of calculating
the NAD/NADH ratio from the equilibrium constant and the intracellular concentrations of
F6P and M1P. Data were verified by thermodynamic analysis. The cytosolic ratio of
NAD/NADH was found to be ~10-fold higher as compared to the same ratio when based on
the whole cell. Under anaerobic conditions however mannitol is formed from M1P implying
that this approach is not yet universally applicable (Costenoble et al. 2003).
A different approach based on a network-embedded thermodynamic analysis later termed
anNET (Zamboni et al. 2008) was used by (Kümmel et al. 2006) to resolve
intracompartmental feasible concentration ranges from cell-averaged metabolome data.
Although these first results are promising there is large open space for the development of
novel strategies combined with appropriate experimental techniques that enable precise
compartment-specific quantification.

3. How can metabolic engineering of S. cerevisiae benefit from quantitative


metabolomics?
In the typical metabolic engineering approach a bunch of new recombinant strains are
designed and created with respect to a particular objective (see Fig. 1) or obtained from
evolutionary adaption. Their new phenotypes are tested by fermentation or conversion
experiments from which the substrate uptake rate and the product pattern in the form of
specific product yields are determined. Results are often applied to FBA for verification.
Intracellular enzyme activities of the introduced reactions as well as of those catalyzing
reactions relevant for the new phenotype are measured from cell-free extracts. This data set
usually provides many valuable details about the production efficiency in terms of
conversion rate (how do intracellular activities of target enzymes compare to the conversion
rate measured) and product selectivity (identification of side-products and oftentimes the
reactions or pathways involved).
Quantitative Metabolomics and Its Application in
Metabolic Engineering of Microbial Cell Factories Exemplified by the Baker’s Yeast 31

So how do metabolomics and more specifically quantitative metabolomics come into play? As
described above the composition of intracellular metabolites together with their levels represent
a direct signature of the physiological state of the cells investigated. Comparing metabolite
profiles of wild-type and mutant strain(s) was often used to identify target reactions limiting the
conversion rate (Hasunuma et al. 2011; Kahar et al. 2011; Klimacek et al. 2010; Kötter and
Ciriacy 1993; Wisselink et al. 2010; Zaldivar et al. 2002) or extract the metabolite pattern
representative for the new phenotype (Canelas et al. 2008b; Devantier et al. 2005; Ding et al.
2010; Hou et al. 2009; Kamei et al. 2011; MacKenzie et al. 2008; Pereira et al. 2011; Raamsdonk et
al. 2001; Ralser et al. 2007; Thorsen et al. 2007; Usaite et al. 2009; Villas-Bôas et al. 2005a; Villas-
Bôas et al. 2005c; Yoshida et al. 2008). Even apparently silent phenotypes of S. cerevisiae single
deletion mutants can be uncovered with respect to the underlying mutation based on the
developed metabolome (Raamsdonk et al. 2001). The rate however at which a compound’s
carbon skeleton is channeled through a certain pathway is directly linked to the level of active
enzymes present and their affinity to the participating reactants as well as to fundamental
thermodynamic laws of the reactions involved. Consequently knowledge about intracellular
concentration of metabolites and enzyme activities combined with thermodynamic and enzyme
kinetic analysis can provide novel and valuable insights into the kinetic organization of the
engineered pathway or even the associated metabolic network which eventually exposes key
regulatory or flux limiting sites. Differently to the general holistic approach usually found in
systems biology pathway analysis in metabolically engineered cells can be reduced in most
cases to the components involved in the new pathway and those connecting this pathway to the
central carbon metabolism (Parachin et al. 2011).

3.1 Thermodynamic pathway analysis


If we are interested in analyzing a pathway or network of pathways on the basis of
thermodynamic rules with the aim to extract pathway or network relevant mechanistic
relationships, knowledge of exact quantitative metabolite data of all reactants involved is
mandatory. On the contrary we can also check quantitative metabolome data with respect to
its thermodynamic consistency (Kümmel et al. 2006) but most importantly we can get first
hits of potential candidate reactions for metabolic engineering within a metabolic network
without any knowledge about enzyme activity and kinetic parameters. Consider the
following reaction

A + B  C + D (1)
The chemical equilibrium constant (Keq) associated with this reaction can be defined
according to the law of mass action as

Keq = cCeqcDeq/(cAeq cBeq) (2)


The superscript eq relates to the concentrations c of reactants at the chemical equilibrium.
The standard Gibbs energy of a chemical reaction (rG0, usually given in J/mol) is related to
Keq by the fundamental relationship

Ns
 rG 0   vi  f G i0   RT ln K eq  (3)
i 1
32 Metabolomics

in which vi and fGi0 correspond to the stoichiometric coefficient of reactant i and to the
standard reaction Gibbs energy of formation of species i at a specified T, P and ionic
strength, respectively. R and T denote the general gas constant (8.314 J/mol/K) and absolute
temperature in Kelvin (K), respectively. The Gibbs energy of formation of a reactant i (fGi)
is further defined by

 fGi = fGi0 + RT ln(ci) (4)


ci in Equation (4) refers to the concentration of reactants involved. The Gibbs energy of a
reaction eventually is described by

Ns Ns
 rG   vi  f Gi0  RT  vi ln ci   r G 0  RT ln Q (5)
i 1 i 1

Q in Equation (5) indicates the reaction quotient cC cD / (cA cB), which is also known under
the term “mass action ratio” that is frequently abbreviated by Γ. Rearranging Equation (4)
yields

 rG = RT {ln(Q/Keq)} (6)


Now the reaction in Equation (2), reading from the left to the right, takes place freely in the
forward direction A,B  C,D at rG < 0 (Keq > Q), is at equilibrium and displays no net flux
when rG = 0 (Keq = Q) and needs support by an external driving force when rG > 0 (Keq <
Q). In other words the reverse reaction is favored under these Q-conditions implying a net
flux in the back direction, C+D  A+B. Equation (6) therefore provides a very convenient
and important expression that permits immediate assignment of the net flux state of a
reaction within a pathway or a large metabolic network provided that Keq and all reactants
participating in the particular reaction are known. The value of Keq is dependent on the
temperature, the ionic strength and the pressure (Alberty 2003). As a consequence only
those Keqs should be applied that were determined under conditions representative for the
cell’s physiological state investigated. In particular temperature, ionic strength and pH are
of considerable interest as pressure can be in most cases assumed as a constant. Thereof only
the temperature during the cultivation experiment, that is usually 30°C for S. cerevisiae, is
known. Unfortunately most Keqs tabulated have been determined at 25°C. For S. cerevisiae
cellular ionic strength and pH are often assumed to be 0.1 M and 7.0, respectively. This may
be sufficient for many thermodynamic network analyses. The validity of these assumptions
however should be tested in any event prior pathway or network analysis. Algorithms are
available that allow to some extent compensation for ionic strength and temperature.
Equilibrium constants can be determined by three different methods based on either in vitro
assay with isolated enzymes, compound-specific standard Gibbs energy of formation (see
Equation (3)) or in vivo experiments that combine FBA with quantitative metabolomics.
In case of using an isolated enzyme a reaction assay is designed such that known
concentrations of substrate(s) and product(s) are solved in a buffer with defined ionic
strength and pH. The reaction is started by the addition of the enzyme and processed at a
certain temperature as long as the equilibrium state is reached. Reactant concentrations are
then analyzed by appropriate techniques and the apparent Keq (K’eq) at a specified pH, ionic
Quantitative Metabolomics and Its Application in
Metabolic Engineering of Microbial Cell Factories Exemplified by the Baker’s Yeast 33

strength and temperature is calculated with respect to the underlying stoichiometry of the
reaction. Values for K’eq are usually applied in the context of network analysis. Importantly
all reactants and the enzyme must be of highest purity and must be stable along the time of
incubation. The composition of the reaction mixture at the equilibrium must not change in
the subsequent component analysis.
When the fGi0 for all reactants participating in the investigated reaction system are known,
the respective values for Keq can be calculated (Alberty 1991). Values for standard Gibbs
energies of formation fGi0 have been tabulated for a number of compounds (Alberty 2003).
Computer programs are available with which one can calculate transformed standard Gibbs
energies of formation (fGi’0) from fGi0 for a specified pH and ionic strength (Alberty 2003;
Zamboni et al. 2008). In addition all possible dissociation forms of a compound are also
lumped into a single reactant in fGi’0. The respective transformed Gibbs energy of
formation for a reactant at a certain concentration (fGi’) is described by

 fGi’ = fGi’0 + RT ln(ci) (7)


Applying fGi’ and fGi’0 instead of fGi and fGi0 in Equations (4) and (5) permit calculation
of K’eq of a particular reaction. Large-scale thermodynamic studies for which availability of
proper K’eqs are crucial often bemoan large uncertainties with which current
thermodynamic data obtained from enzyme assays or based on Gibbs energies are afflicted
and therefore not sufficient for data analysis.
A way to circumvent these obstacles was introduced by (Canelas et al. 2011) who
developed a new method to derive apparent equilibrium constants under real in vivo
conditions. The basic idea of this work was based upon that the rate v of a certain reaction
is directly dependent on the maximal turnover number Vmax, the kinetic properties () of
the enzyme catalyzing this reaction and on the net flux state of the reaction in relation to
the respective equilibrium (1-Q/K’eq). That is v = Vmax(1-Q/K’eq). S. cerevisiae was
cultivated at 30°C in chemostats under aerobic conditions and glucose (7.5 g/L) as the
sole carbon source in standard mineral medium at 32 different dilution rates spanning a
wide range of specific growth rates (0.03 to 0.29 h-1) and as a consequence substrate
conversion rates. Extracellular and intracellular metabolites were quantified at each
dilution rate. Specific substrate and product fluxes given in mmol/gCDW/h were applied
to a FBA based on a stoichiometric model and stoichiometric fluxes (v) for each reaction of
the model network were calculated. Mass ratio coefficients Q of 27 network reactions
were calculated from quantitative metabolite data at each v. From plots of Q vs. v the
authors were able to extract values for K’eqs. It could be shown that Q of reactions
operating close to the equilibrium state display a negative linear dependency of v. Values
of K’eqs representing the thermodynamic equilibrium in the cell were determined by
linear extrapolation to the y-axis (v = 0 and K’eq = Q) under the assumption that Vmax and
kinetic properties of enzymes involved are not dependent on v. Intracellular K’eqs
obtained differed in part significantly (up to a factor of 9) from their in vitro determined
counterparts implying that reaction conditions in vivo (pH and ionic strength) deviate
from that specified in in vitro assays or by the in silico approach. The big advantage of
this approach is that in vivo K’eqs can be accurately determined without knowing
anything about the intracellular pH and ionic strength. However only reactions located at
or close to the equilibrium state are addressable.
34 Metabolomics

Combining quantitative metabolomics data with thermodynamic rules on reactions


constituting an operating metabolic network enables one to map reactions according to their
location with respect to K’eq (Crabtree et al. 1997; Klimacek et al. 2010; Kümmel et al. 2006;
Wang et al. 2004). That is close to or far away from equilibrium. A reaction reaches K’eq
when the activity of an enzyme catalyzing a reaction downstream is low. On the other hand
reactions located far away from K’eq are often catalyzed by enzymes that have regulatory
functions in the cell or by flux limiting enzymes representing potential targets for strain
improvement (Klimacek et al. 2010). Differently to the relative location of a particular
reaction to its equilibrium, the sign (plus or minus) of rG’ or whether Q > or < K’eq gives
important information about the direction of the reaction or of an entire pathway.
Information about flux directions can be readily implemented as further restrictions into a
stoichiometric network to increase reliability of flux distributions (Hoppe et al. 2007;
Kümmel et al. 2006).

3.2 Metabolic control analysis based on quantitative metabolomics in metabolic


engineering of S. cerevisiae
Identification of those reactions exerting significant control of flux through a particular
pathway is crucial to develop strategies for flux improvement. As mentioned above
reactions suspected in flux limitation can be elucidated elegantly by thermodynamic
analysis. Once identified reactions can be further analyzed with respect to principles of
MCA. The concept of MCA was developed and introduced by (Heinrich and Rapoport 1974;
Kacser and Burns 1973) and is comprehensively described in (Stephanopoulos et al. 1998).
MCA strictly applies only to steady-state conditions. To unravel whether the amount of
enzyme or of a reactant participating in the reaction catalyzed by this enzyme contribute to
flux limitation through this particular reaction two terms have been defined, namely the flux
control coefficient (FCC) and the elasticity coefficient (). The FCC defines the relative
change in the steady-state flux resulting from an infinitesimal change in the activity of an
enzyme of the pathway divided by the relative change of the enzyme activity, represented
by the relationship FCC = E dJ / (J dE), in which E and J stand for the concentration or
activity of the enzyme and the steady-state flux, respectively. The  is intrinsically linked to
the inherent enzyme kinetic properties and is defined by the ratio of the relative change in
the reaction rate resulted by an infinitesimal change in the metabolite concentration and can
be described by  = cj vi / (vi cj), where cj and vi represent the concentration of reactant j
and the reaction rate of enzyme i. FCC and  are related by the flux-control connectivity
theorem (Kacser and Burns 1973).

L
 FCCiJ ci j
0 (8)
i 1

Equation (8) implies that large elasticities – concentration of reactant exerts large control on
the enzyme reaction rate – are associated with small FCCs – the overall flux through the
pathway is not very dependent on the enzyme activity – and vice versa. Consequently
knowledge of either of these coefficient is only required. FCC can be determined by
increasing the amount of active enzyme for example by expressing this particular enzyme at
different expression levels and measuring the change in flux through the pathway. The level
of enzyme present can be judged by determining its specific activity in cell-free extracts. If
Quantitative Metabolomics and Its Application in
Metabolic Engineering of Microbial Cell Factories Exemplified by the Baker’s Yeast 35

the specific activity (µmol/mgprotein/min) for the isolated enzyme, the cellular protein
content (mgprotein/gCDW) and the specific cell volume are known then the molar
concentration of this enzyme can be calculated. Enzyme levels can be also determined by
proteomics techniques and to some extent extrapolated from transcriptome data.
Alternatively suitable antibodies that selectively bind to the target enzymes or fusion of an
indicator peptide or protein tag to the enzyme to be analyzed permitting quantification
either directly in vivo (GFP) or in vitro after separation of the target enzyme by affinity
chromatography (e.g. His-tag, strep-tag) can be used for determining intracellular enzyme
concentrations. If rate equations and associated kinetic parameters of all enzymes involved
and at least their relative activity levels are known mathematical models can be applied to
estimate FCCs and 's. Rate equations based on the steady-state or rapid equilibrium
assumption for enzyme catalyzed reactions are comprehensively summarized in (Segel
1993). However kinetic parameters typically used in this approach are determined by in
vitro assays and it has been shown that in vitro generated data can significantly differ from
those observed in vivo (Aragón and Sánchez 1985; Mauch et al. 2000; Reuss et al. 2007). This
discrepancy is most likely due to the incomplete knowledge of the cellular composition and
associated enzyme metabolite interactions as well as the difficulty to analyze enzymes under
in vivo-like conditions at the lab bench. A promising step forward towards generation of
more reliable in vivo-like in vitro enzyme kinetic data was reported recently (van Eunen et
al. 2010). The authors suggested to measure enzyme activities in a buffered reaction mixture
that simulates the intracellular cellular medium composition of S. cerevisiae.
Using the non-linear lin-log formulation developed for metabolic network analysis by
(Visser and Heijnen 2003) FCC and  can be estimated without any prior knowledge of
enzyme-specific kinetic parameters (Visser and Heijnen 2002). In this approach the reaction
rate is a nonlinear function of metabolite concentrations and is proportional to enzyme
levels. lin-log kinetics is suited when large perturbations on the systems are analyzed.
Furthermore statistical evaluation of parameter estimates is simplified (Wu et al. 2004).
Nevertheless it is an approximation that fits to fundamental enzyme-reactant properties
only in a certain range of perturbation (Wu et al. 2004).

3.3 Substrate promiscuous enzymes


Enzymes that have evolved relaxed or broad substrate specificity are substrate promiscuous
(Hult and Berglund 2007). The presence of substrate promiscuous enzymes in a metabolic
network considerably aggravates accurate network formulation and analysis if the accurate
flux partition between the individual substrates is not known. This problem is further
enhanced when the utilized substrates are highly connected within the network by other
(substrate promiscuous) reactions. This is possibly best exemplified by the coenzyme
promiscuous enzyme xylose reductase (XR) from Candida tenuis (Petschacher et al. 2005). To
enable metabolic integration of xylose by S. cerevisiae XR and xylitol dehydrogenase XDH
have to be integrated (Petschacher and Nidetzky 2008). Different to XDH which is strictly
dependent on NAD(H) (Nidetzky et al. 2003), XR can oxidize both NADH and NADPH.
These coenzymes in turn form the redox state of a cell and are important currency
metabolites in all catabolic and anabolic pathways of the cell. Hence, keeping the redox state
with respect to anabolism and catabolism balanced is crucial for the cell to stay alive. It was
found based on stoichiometric considerations that the NADPH usage of XR is in strong
36 Metabolomics

correlation with by-product formation in the form of xylitol, the reaction product of XR and
the substrate of XDH in the subsequent reaction (van Maris et al. 2007). Usage of up to (0-
52)% NADPH by XR was compatible with a genome-scale metabolic network (Krahulec et
al. 2010). Detailed kinetic analysis from in vitro studies showed that the XR almost
exclusively utilizes NADPH in terms of catalytic efficiency (kcat/Km,coenzyme) and the
selectivity parameter Rsel (kcat/(Ki,coenzymeKm,xylose)) which are widely used as marker
parameter for coenzyme discrimination. Based on this data even a rough estimation of the
coenzyme usage of XR in the cell is not possible. To solve the coenzyme usage riddle of XR
we determined intracellular concentrations of NADH and NADPH (Klimacek et al. 2010)
and integrated this information together with the relevant kinetic parameters (Petschacher
et al. 2005) into the mechanistically appropriate enzyme kinetic rate expression (Banta et al.
2002; Petschacher and Nidetzky 2005). A balanced coenzyme usage perfectly in line with
physiology observed was obtained for the XR (Klimacek et al. 2010). Information about the
correct flux partition of a particular substrate promiscuous enzyme can be implemented as
further restrictions into a stoichiometric network to increase reliability of flux distributions.
Furthermore this approach could be successfully applied on a series of wild-type and
mutant forms of XR to predict reliably formation of xylitol (Krahulec et al. 2011).

4. Conclusions
Quantitative metabolomics is especially suited to help identifying key sites limiting an
engineered metabolic route either within the created pathway but also apart from it. State-
of-the-art protocols for sample work-up and LC-MS and GC-MS analysis permit absolute
quantification of metabolites from S. cerevisiae cells provided that a U-13C-labled IS is
applied. Quantitative data in turn are indispensible for reliable pathway and network
analysis in the form of a thermodynamic analysis, MCA or kinetic modelling. In
combination with other omics techniques it represents a powerful tool to create designer
microbial cell factories exposing improved or novel phenotypes.

5. Acknowledgement
Financial support from the Austrian Science Fund FWF (J2698) is gratefully acknowledged.

6. References
Abbott DA, Zelle RM, Pronk JT, van Maris AJ. 2009. Metabolic engineering of Saccharomyces
cerevisiae for production of carboxylic acids: current status and challenges. FEMS
Yeast Res 9(8):1123-36.
Alberty RA. 1991. Equilibrium compositions of solutions of biochemical species and heats of
biochemical reactions. Proc Natl Acad Sci U S A 88(8):3268-71.
Alberty RA. 2003. Thermodynamics of biochemical reactions. Alberty RA, editor. New
Jersey: John Wiley & Sons, Inc.
Alivisatos SG, Ungar F, Abraham G. 1964. Non-Enzymatic Interactions of Reduced
Coenzyme I with Inorganic Phosphate and Certain Other Anions. Nature 203:973-5.
Aragón JJ, Sánchez V. 1985. Enzyme concentration affects the allosteric behavior of yeast
phosphofructokinase. Biochem Biophys Res Commun 131(2):849-55.
Quantitative Metabolomics and Its Application in
Metabolic Engineering of Microbial Cell Factories Exemplified by the Baker’s Yeast 37

Asadollahi MA, Maury J, Patil KR, Schalk M, Clark A, Nielsen J. 2009. Enhancing
sesquiterpene production in Saccharomyces cerevisiae through in silico driven
metabolic engineering. Metab Eng 11(6):328-34.
Asadollahi MA, Maury J, Schalk M, Clark A, Nielsen J. 2010. Enhancement of farnesyl
diphosphate pool as direct precursor of sesquiterpenes through metabolic
engineering of the mevalonate pathway in Saccharomyces cerevisiae. Biotechnol
Bioeng 106(1):86-96.
Bailey JE, Sburlati A, Hatzimanikatis V, Lee K, Renner WA, Tsai PS. 2002. Inverse metabolic
engineering: a strategy for directed genetic engineering of useful phenotypes.
Biotechnol Bioeng 79(5):568-79.
Banta S, Boston M, Jarnagin A, Anderson S. 2002. Mathematical modeling of in vitro
enzymatic production of 2-Keto-L-gulonic acid using NAD(H) or NADP(H) as
cofactors. Metab Eng 4(4):273-84.
Bengtsson O, Jeppsson M, Sonderegger M, Parachin NS, Sauer U, Hahn-Hagerdal B, Gorwa-
Grauslund MF. 2008. Identification of common traits in improved xylose-growing
Saccharomyces cerevisiae for inverse metabolic engineering. Yeast 25(11):835-47.
Bettiga M, Gorwa-Grauslund MF, Hahn-Hägerdal B. 2010. Metabolic engineering in yeast.
In: Smolke CD, editor. The metabolic pathway engineering handbook.
Fundamentals. Boca Raton: CRC Press. p 1 - 48.
Brauer MJ, Huttenhower C, Airoldi EM, Rosenstein R, Matese JC, Gresham D, Boer VM,
Troyanskaya OG, Botstein D. 2008. Coordination of growth rate, cell cycle, stress
response, and metabolic activity in yeast. Mol Biol Cell 19(1):352-67.
Bro C, Knudsen S, Regenberg B, Olsson L, Nielsen J. 2005. Improvement of galactose uptake
in Saccharomyces cerevisiae through overexpression of phosphoglucomutase:
example of transcript analysis as a tool in inverse metabolic engineering. Appl
Environ Microbiol 71(11):6465-72.
Bro C, Regenberg B, Förster J, Nielsen J. 2006. In silico aided metabolic engineering of
Saccharomyces cerevisiae for improved bioethanol production. Metab Eng 8(2):102-11.
Buescher JM, Moco S, Sauer U, Zamboni N. 2010. Ultrahigh performance liquid
chromatography-tandem mass spectrometry method for fast and robust
quantification of anionic and aromatic metabolites. Anal Chem 82(11):4403-12.
Büscher JM, Czernik D, Ewald JC, Sauer U, Zamboni N. 2009. Cross-platform comparison of
methods for quantitative metabolomics of primary metabolism. Anal Chem
81(6):2135-43.
Cakar ZP, Alkim C, Turanli B, Tokman N, Akman S, Sarikaya M, Tamerler C, Benbadis L,
François JM. 2009. Isolation of cobalt hyper-resistant mutants of Saccharomyces
cerevisiae by in vivo evolutionary engineering approach. J Biotechnol 143(2):130-8.
Cakar ZP, Seker UO, Tamerler C, Sonderegger M, Sauer U. 2005. Evolutionary engineering
of multiple-stress resistant Saccharomyces cerevisiae. FEMS Yeast Res 5(6-7):569-78.
Canelas AB, Ras C, Ten Pierick A, van Dam JC, Heijnen JJ, van Gulik W. 2008a. Leakage-free
rapid quenching technique for yeast metabolomics. Metabolomics 4:226-239.
Canelas AB, Ras C, ten Pierick A, van Gulik WM, Heijnen JJ. 2011. An in vivo data-driven
framework for classification and quantification of enzyme kinetics and
determination of apparent thermodynamic data. Metab Eng 13(3):294-306.
Canelas AB, ten Pierick A, Ras C, Seifar RM, van Dam JC, van Gulik WM, Heijnen JJ. 2009.
Quantitative evaluation of intracellular metabolite extraction techniques for yeast
metabolomics. Anal Chem 81(17):7379-89.
38 Metabolomics

Canelas AB, van Gulik WM, Heijnen JJ. 2008b. Determination of the cytosolic free
NAD/NADH ratio in Saccharomyces cerevisiae under steady-state and highly
dynamic conditions. Biotechnol Bioeng 100(4):734-43.
Castrillo JI, Oliver SG. 2005. Towards integrative functional genomics using yeast as a
reference model. In: Vaidyanathan S, Harrigan GG, Goodacre R, editors.
Metabolome analysis: Strategies for systems biology. Berlin: Springer-Verlag. p 9 -
30.
Castrillo JI, Oliver SG. 2006. Metabolomics and systems biology in Saccharomyces
cerevisiae. In: Brown AJP, editor. Fungal Genomics. Berlin: Springer-Verlag. p 3 - 7.
Chaykin S. 1967. Nicotinamide coenzymes. Annu Rev Biochem 36:149-70.
Cipollina C, van den Brink J, Daran-Lapujade P, Pronk JT, Vai M, de Winde JH. 2008.
Revisiting the role of yeast Sfp1 in ribosome biogenesis and cell size control: a
chemostat study. Microbiology 154(Pt 1):337-46.
Ciriacy M, Breitenbach I. 1979. Physiological effects of seven different blocks in glycolysis in
Saccharomyces cerevisiae. J Bacteriol 139(1):152-60.
Costenoble R, Adler L, Niklasson C, Lidén G. 2003. Engineering of the metabolism of
Saccharomyces cerevisiae for anaerobic production of mannitol. FEMS Yeast Res
3(1):17-25.
Crabtree B, Newsholme EA, Reppas NB. 1997. Principles of regulation and control in
biochemistry: a paradigmatic, flux-oriented approach. In: Hoffman JF, Jamieson JD,
editors. Handbook of Physiology. New York, U.S.A.: Oxford University Press. p
117-180.
Crabtree HG. 1928. The carbohydrate metabolism of certain pathological overgrowths.
Biochem J 22(5):1289-98.
Csuk R. 1991. Baker's yeast mediated transformations on organic chemistry. Chem Rev 91:49
- 97.
Cvijovic M, Bordel S, Nielsen J. 2010. Mathematical models of cell factories: moving towards
the core of industrial biotechnology. Microb Biotechnol 4(5):572-84.
de Koning W, van Dam K. 1992. A method for the determination of changes of glycolytic
metabolites in yeast on a subsecond time scale using extraction at neutral pH. Anal
Biochem 204(1):118-23.
Dejong JM, Liu Y, Bollon AP, Long RM, Jennewein S, Williams D, Croteau RB. 2006. Genetic
engineering of taxol biosynthetic genes in Saccharomyces cerevisiae. Biotechnol
Bioeng 93(2):212-24.
den Hollander JA, Ugurbil K, Brown TR, Shulman RG. 1981. Phosphorus-31 nuclear
magnetic resonance studies of the effect of oxygen upon glycolysis in yeast.
Biochemistry 20(20):5871-80.
Devantier R, Scheithauer B, Villas-Bôas SG, Pedersen S, Olsson L. 2005. Metabolite profiling
for analysis of yeast stress response during very high gravity ethanol
fermentations. Biotechnol Bioeng 90(6):703-14.
Ding M-Z, Zhou X, Yuan Y-J. 2010. Metabolome profiling reveals adaptive evolution of
Saccharomyces cerevisiae during repeated vacuum fermentations. Metabolomics 6:42
- 55.
Dinh TN, Nagahisa K, Hirasawa T, Furusawa C, Shimizu H. 2008. Adaptation of
Saccharomyces cerevisiae cells to high ethanol concentration and changes in fatty acid
composition of membrane and cell size. PLoS One 3(7):e2623.
Quantitative Metabolomics and Its Application in
Metabolic Engineering of Microbial Cell Factories Exemplified by the Baker’s Yeast 39

Ditzelmüller G, Wöhrer W, Kubicek CP, Röhr M. 1983. Nucleotide pools of growing,


synchronized and stressed cultures of Saccharomyces cerevisiae. Arch Microbiol
135(1):63-7.
Domingues L, Guimarães PM, Oliveira C. 2010. Metabolic engineering of Saccharomyces
cerevisiae for lactose/whey fermentation. Bioeng Bugs 1(3):164-171.
Förster J, Famili I, Fu P, Palsson BØ, Nielsen J. 2003. Genome-scale reconstruction of the
Saccharomyces cerevisiae metabolic network. Genome Res 13(2):244-53.
Garcia Sanchez R, Karhumaa K, Fonseca C, Sànchez Nogué V, Almeida JR, Larsson CU,
Bengtsson O, Bettiga M, Hahn-Hägerdal B, Gorwa-Grauslund MF. 2010. Improved
xylose and arabinose utilization by an industrial recombinant Saccharomyces
cerevisiae strain using evolutionary engineering. Biotechnol Biofuels 3:13.
Gonzalez B, François J, Renaud M. 1997. A rapid and reliable method for metabolite
extraction in yeast using boiling buffered ethanol. Yeast 13(14):1347-55.
Grosz R, Stephanopoulos G. 1990. Physiological, biochemical, and mathematical studies of
micro-aerobic continuous ethanol fermentation by Saccharomyces cerevisiae. II:
intracellular metabolite and enzyme assays at steady state chemostat cultures.
Biotechnol Bioeng 36(10):1020-9.
Hasunuma T, Sanda T, Yamada R, Yoshimura K, Ishii J, Kondo A. 2011. Metabolic pathway
engineering based on metabolomics confers acetic and formic acid tolerance to a
recombinant xylose-fermenting strain of Saccharomyces cerevisiae. Microb Cell Fact
10(1):2.
Heinrich R, Rapoport TA. 1974. A linear steady-state treatment of enzymatic chains. General
properties, control and effector strength. Eur J Biochem 42(1):89-95.
Hong ME, Lee KS, Yu BJ, Sung YJ, Park SM, Koo HM, Kweon DH, Park JC, Jin YS. 2010.
Identification of gene targets eliciting improved alcohol tolerance in Saccharomyces
cerevisiae through inverse metabolic engineering. J Biotechnol 149(1-2):52-9.
Hoppe A, Hoffmann S, Holzhutter HG. 2007. Including metabolite concentrations into flux
balance analysis: thermodynamic realizability as a constraint on flux distributions
in metabolic networks. BMC Syst Biol 1:23.
Hou J, Lages NF, Oldiges M, Vemuri GN. 2009. Metabolic impact of redox cofactor
perturbations in Saccharomyces cerevisiae. Metab Eng 11(4-5):253-61.
Hult K, Berglund P. 2007. Enzyme promiscuity: mechanism and applications. Trends
Biotechnol 25(5):231-8.
Ishida N, Saitoh S, Ohnishi T, Tokuhiro K, Nagamori E, Kitamoto K, Takahashi H. 2006.
Metabolic engineering of Saccharomyces cerevisiae for efficient production of pure L-
(+)-lactic acid. Appl Biochem Biotechnol 129-132:795-807.
Jackson BE, Hart-Wells EA, Matsuda SP. 2003. Metabolic engineering to produce
sesquiterpenes in yeast. Org Lett 5(10):1629-32.
Jeffries TW, Jin YS. 2004. Metabolic engineering for improved fermentation of pentoses by
yeasts. Appl Microbiol Biotechnol 63(5):495-509.
Jin YS, Alper H, Yang YT, Stephanopoulos G. 2005. Improvement of xylose uptake and
ethanol production in recombinant Saccharomyces cerevisiae through an inverse
metabolic engineering approach. Appl Environ Microbiol 71(12):8249-56.
Johnston GC. 1977. Cell size and budding during starvation of the yeast Saccharomyces
cerevisiae. J Bacteriol 132(2):738-9.
Johnston GC, Ehrhardt CW, Lorincz A, Carter BL. 1979. Regulation of cell size in the yeast
Saccharomyces cerevisiae. J Bacteriol 137(1):1-5.
Kacser H, Burns JA. 1973. The control of flux. Symp Soc Exp Biol 27:65 - 104.
40 Metabolomics

Kahar P, Taku K, Tanaka S. 2011. Enhancement of xylose uptake in 2-deoxyglucose tolerant


mutant of Saccharomyces cerevisiae. J Biosci Bioeng 111(5):557 - 563.
Kamei Y, Tamura T, Yoshida R, Ohta S, Fukusaki E, Mukai Y. 2011. GABA metabolism
pathway genes, UGA1 and GAD1, regulate replicative lifespan in Saccharomyces
cerevisiae. Biochem Biophys Res Commun 407(1):185-90.
Kjeldsen T. 2000. Yeast secretory expression of insulin precursors. Appl Microbiol
Biotechnol 54(3):277-86.
Klimacek M, Krahulec S, Sauer U, Nidetzky B. 2010. Limitations in xylose-fermenting
Saccharomyces cerevisiae, made evident through comprehensive metabolite profiling
and thermodynamic analysis. Appl Environ Microbiol 76(22):7566-74.
Kötter P, Ciriacy M. 1993. Xylose fermentation by Saccharomyces cerevisiae. Appl Microbiol
Biotechnol 38:776 - 783.
Krahulec S, Klimacek M, Nidetzky B. 2009. Engineering of a matched pair of xylose
reductase and xylitol dehydrogenase for xylose fermentation by Saccharomyces
cerevisiae. Biotechnol J 4(5):684-94.
Krahulec S, Klimacek M, Nidetzky B. 2011. Analysis and prediction of the physiological
effects of altered coenzyme specificity in xylose reductase and xylitol
dehydrogenase during xylose fermentation by Saccharomyces cerevisiae. J Biotechnol
in press.
Krahulec S, Petschacher B, Wallner M, Longus K, Klimacek M, Nidetzky B. 2010.
Fermentation of mixed glucose-xylose substrates by engineered strains of
Saccharomyces cerevisiae: role of the coenzyme specificity of xylose reductase, and
effect of glucose on xylose utilization. Microb Cell Fact 9:16.
Kubota S, Takeo I, Kume K, Kanai M, Shitamukai A, Mizunuma M, Miyakawa T, Shimoi H,
Iefuji H, Hirata D. 2004. Effect of ethanol on cell growth of budding yeast: genes
that are important for cell growth in the presence of ethanol. Biosci Biotechnol
Biochem 68(4):968-72.
Kümmel A, Panke S, Heinemann M. 2006. Putative regulatory sites unraveled by network-
embedded thermodynamic analysis of metabolome data. Mol Syst Biol 2:2006 0034.
Kuroda S, Miyazaki T, Otaka S, Fujisawa Y. 1993. Saccharomyces cerevisiae can release
hepatitis B virus surface antigen (HBsAg) particles into the medium by its secretory
apparatus. Appl Microbiol Biotechnol 40(2-3):333-40.
Kuyper M, Toirkens MJ, Diderich JA, Winkler AA, van Dijken JP, Pronk JT. 2005.
Evolutionary engineering of mixed-sugar utilization by a xylose-fermenting
Saccharomyces cerevisiae strain. FEMS Yeast Res 5(10):925-34.
Lee KS, Hong ME, Jung SC, Ha SJ, Yu BJ, Koo HM, Park SM, Seo JH, Kweon DH, Park JC
and others. 2010. Improved galactose fermentation of Saccharomyces cerevisiae
through inverse metabolic engineering. Biotechnol Bioeng 108(3):621-31.
Lord PG, Wheals AE. 1981. Variability in individual cell cycles of Saccharomyces cerevisiae. J
Cell Sci 50:361-76.
Lorincz A, Carter BL. 1979. Control of cell size at bud initiation in Saccharomyces cerevisiae. J
Gen Microbiol 113:287 - 295.
Lowry OH, Passonneau JV, Rock MK. 1961. The stability of pyridine nucleotides. J Biol
Chem 236:2756-9.
Lu W, Clasquin MF, Melamud E, Amador-Noguez D, Caudy AA, Rabinowitz JD. 2010.
Metabolomic analysis via reversed-phase ion-pairing liquid chromatography
coupled to a stand alone orbitrap mass spectrometer. Anal Chem 82(8):3212-21.
Quantitative Metabolomics and Its Application in
Metabolic Engineering of Microbial Cell Factories Exemplified by the Baker’s Yeast 41

MacKenzie DA, Defernez M, Dunn WB, Brown M, Fuller LJ, de Herrera SR, Günther A,
James SA, Eagles J, Philo M and others. 2008. Relatedness of medically important
strains of Saccharomyces cerevisiae as revealed by phylogenetics and metabolomics.
Yeast 25(7):501-12.
Madsen KM, Udatha GD, Semba S, Otero JM, Koetter P, Nielsen J, Ebizuka Y, Kushiro T,
Panagiotou G. 2011. Linking genotype and phenotype of Saccharomyces cerevisiae
strains reveals metabolic engineering targets and leads to triterpene hyper-
producers. PLoS One 6(3):e14763.
Mashego MR, Wu L, Van Dam JC, Ras C, Vinke JL, Van Winden WA, Van Gulik WM,
Heijnen JJ. 2004. MIRACLE: mass isotopomer ratio analysis of U-13C-labeled
extracts. A new method for accurate quantification of changes in concentrations of
intracellular metabolites. Biotechnol Bioeng 85(6):620-8.
Mauch K, Vaseghi S, Reuss M. 2000. Quantitative analysis of metabolic signalling pathways
in Saccharomyces cerevisiae. In: Schügerl K, Bellgardt KH, editors. Bioreaction
engineering. Berlin: Springer Verlag. p 435 - 477.
Mountain HA, Sudbery PE. 1990. The relationship of growth rate and catabolite repression with
WHI2 expression and cell size in Saccharomyces cerevisiae. J Gen Microbiol 136(4):733-7.
Mutka SC, Bondi SM, Carney JR, Da Silva NA, Kealey JT. 2006. Metabolic pathway
engineering for complex polyketide biosynthesis in Saccharomyces cerevisiae. FEMS
Yeast Res 6(1):40-7.
Navon G, Shulman RG, Yamane T, Eccleshall TR, Lam KB, Baronofsky JJ, Marmur J. 1979.
Phosphorus-31 nuclear magnetic resonance studies of wild-type and glycolytic
pathway mutants of Saccharomyces cerevisiae. Biochemistry 18(21):4487-99.
Nevoigt E. 2008. Progress in metabolic engineering of Saccharomyces cerevisiae. Microbiol Mol
Biol Rev 72(3):379-412.
Nidetzky B, Helmer H, Klimacek M, Lunzer R, Mayer G. 2003. Characterization of
recombinant xylitol dehydrogenase from Galactocandida mastotermitis expressed in
Escherichia coli. Chem. Biol. Interact. 143-144:533-542.
Nielsen J. 2007. Metabolomics in functional genomics and systems biology. In: Villas-Bôas
SG, Roessner U, Hansen MAE, Smedsgaard J, Nielsen J, editors. Metabolome
analysis: An Introduction. Hoboken: John Wiley & Sons, Inc. p 3 - 14.
Nielsen J, Jewett MC. 2008. Impact of systems biology on metabolic engineering of
Saccharomyces cerevisiae. FEMS Yeast Res 8(1):122-31.
Nissen TL, Hamann CW, Kielland-Brandt MC, Nielsen J, Villadsen J. 2000a. Anaerobic and
aerobic batch cultivations of Saccharomyces cerevisiae mutants impaired in glycerol
synthesis. Yeast 16(5):463-74.
Nissen TL, Kielland-Brandt MC, Nielsen J, Villadsen J. 2000b. Optimization of ethanol
production in Saccharomyces cerevisiae by metabolic engineering of the ammonium
assimilation. Metab Eng 2(1):69-77.
Parachin NS, Bergdahl B, van Niel EW, Gorwa-Grauslund MF. 2011. Kinetic modelling
reveals current limitations in the production of ethanol from xylose by recombinant
Saccharomyces cerevisiae. Metab Eng in press.
Pereira FB, Guimarães PMR, Teixeira JA, Domingues L. 2011. Robust industrial
Saccharomyces cerevisiae strains for very high gravity bio-ethanol fermentations. J
Biosci Bioeng 112(2):130 - 136.
Petschacher B, Leitgeb S, Kavanagh KL, Wilson DK, Nidetzky B. 2005. The coenzyme
specificity of Candida tenuis xylose reductase (AKR2B5) explored by site-directed
mutagenesis and X-ray crystallography. Biochem. J. 385(Pt 1):75-83.
42 Metabolomics

Petschacher B, Nidetzky B. 2005. Engineering Candida tenuis Xylose reductase for improved
utilization of NADH: antagonistic effects of multiple side chain replacements and
performance of site-directed mutants under simulated in vivo conditions. Appl.
Environ. Microbiol. 71(10):6390-6393.
Petschacher B, Nidetzky B. 2008. Altering the coenzyme preference of xylose reductase to
favor utilization of NADH enhances ethanol yield from xylose in a metabolically
engineered strain of Saccharomyces cerevisiae. Microb Cell Fact 7:9.
Pirkov I, Albers E, Norbeck J, Larsson C. 2008. Ethylene production by metabolic
engineering of the yeast Saccharomyces cerevisiae. Metab Eng 10(5):276-80.
Porro D, Brambilla L, Alberghina L. 2003. Glucose metabolism and cell size in continuous
cultures of Saccharomyces cerevisiae. FEMS Microbiol Lett 229(2):165-71.
Raab AM, Gebhardt G, Bolotina N, Weuster-Botz D, Lang C. 2010. Metabolic engineering of
Saccharomyces cerevisiae for the biotechnological production of succinic acid. Metab
Eng 12(6):518-25.
Raamsdonk LM, Teusink B, Broadhurst D, Zhang N, Hayes A, Walsh MC, Berden JA,
Brindle KM, Kell DB, Rowland JJ and others. 2001. A functional genomics strategy
that uses metabolome data to reveal the phenotype of silent mutations. Nat
Biotechnol 19(1):45-50.
Racker E. 1974. History of the Pasteur effect and its pathobiology. Mol Cell Biochem 5(1-
2):17-23.
Ralser M, Wamelink MM, Kowald A, Gerisch B, Heeren G, Struys EA, Klipp E, Jakobs C,
Breitenbach M, Lehrach H and others. 2007. Dynamic rerouting of the carbohydrate
flux is key to counteracting oxidative stress. J Biol 6(4):10.
Reuss M, Aguilera-Vázques L, Mauch K. 2007. Reconstruction of dynamic network models
from metabolite measurements. In: Nielsen J, Jewett MC, editors. Metabolomics.
Berlin: Springer-Verlag. p 97 - 127.
Rizzi M, Baltes M, Theobald U, Reuss M. 1997. In vivo analysis of metabolic dynamics in
Saccharomyces cerevisiae: II. Mathematical model. Biotechnol Bioeng 55(4):592-608.
Rocha I, Maia P, Evangelista P, Vilaca P, Soares S, Pinto JP, Nielsen J, Patil KR, Ferreira EC,
Rocha M. OptFlux: an open-source software platform for in silico metabolic
engineering. BMC Syst Biol 4:45.
Segel IH. 1993. Enzyme Kinetics - Behavior and analysis of rapid equilibrium and steady-
state enzyme systems. New York: Wiley interscience.
Selvarajoo K, Arjunan SNV, Tomita M. 2010. In silico models for metabolic systems
engineering. In: Smolke CD, editor. The metabolic pathway engineering handbook.
Tools and applications. Boca Raton: CRC Press. p 1 - 22.
Shanks JV, Bailey JE. 1988. Estimation of intracellular sugar phosphate concentrations in
Saccharomyces cerevisiae using 31P nuclear magnetic resonance spectroscopy.
Biotechnol Bioeng 32(9):1138-52.
Sonderegger M, Sauer U. 2003. Evolutionary engineering of Saccharomyces cerevisiae for
anaerobic growth on xylose. Appl Environ Microbiol 69(4):1990-8.
Steen EJ, Chan R, Prasad N, Myers S, Petzold CJ, Redding A, Ouellet M, Keasling JD. 2008.
Metabolic engineering of Saccharomyces cerevisiae for the production of n-butanol.
Microb Cell Fact 7:36.
Steinle A, Bergander K, Steinbüchel A. 2009. Metabolic engineering of Saccharomyces
cerevisiae for production of novel cyanophycins with an extended range of
constituent amino acids. Appl Environ Microbiol 75(11):3437-46.
Quantitative Metabolomics and Its Application in
Metabolic Engineering of Microbial Cell Factories Exemplified by the Baker’s Yeast 43

Stephanopoulos GN, Aristidou A, Nielsen J. 1998. Metabolic engineering, principles and


methodologies. San Diego: Academic Press.
Tamaki H, Yun CW, Mizutani T, Tsuzuki T, Takagi Y, Shinozaki M, Kodama Y, Shirahige K,
Kumagai H. 2005. Glucose-dependent cell size is regulated by a G protein-coupled
receptor system in yeast Saccharomyces cerevisiae. Genes Cells 10(3):193-206.
Theobald U, Mailinger W, Baltes M, Rizzi M, Reuss M. 1997. In vivo analysis of metabolic
dynamics in Saccharomyces cerevisiae : I. Experimental observations. Biotechnol
Bioeng 55(2):305-16.
Thorsen M, Lagniel G, Kristiansson E, Junot C, Nerman O, Labarre J, Tamás MJ. 2007.
Quantitative transcriptome, proteome, and sulfur metabolite profiling of the
Saccharomyces cerevisiae response to arsenite. Physiol Genomics 30(1):35-43.
Toivari MH, Ruohonen L, Miasnikov AN, Richard P, Penttilä M. 2007. Metabolic
engineering of Saccharomyces cerevisiae for conversion of D-glucose to xylitol and
other five-carbon sugars and sugar alcohols. Appl Environ Microbiol 73(17):5471-6.
Trantas E, Panopoulos N, Ververidis F. 2009. Metabolic engineering of the complete
pathway leading to heterologous biosynthesis of various flavonoids and stilbenoids
in Saccharomyces cerevisiae. Metab Eng 11(6):355-66.
Tyson CB, Lord PG, Wheals AE. 1979. Dependency of size of Saccharomyces cerevisiae cells on
growth rate. J Bacteriol 138(1):92-8.
Ukibe K, Hashida K, Yoshida N, Takagi H. 2009. Metabolic engineering of Saccharomyces
cerevisiae for astaxanthin production and oxidative stress tolerance. Appl Environ
Microbiol 75(22):7205-11.
Usaite R, Jewett MC, Oliveira AP, Yates JR, 3rd, Olsson L, Nielsen J. 2009. Reconstruction of
the yeast Snf1 kinase regulatory network reveals its role as a global energy
regulator. Mol Syst Biol 5:319.
van Eunen K, Bouwman J, Daran-Lapujade P, Postmus J, Canelas AB, Mensonides FI, Orij R,
Tuzun I, van den Brink J, Smits GJ and others. 2010. Measuring enzyme activities
under standardized in vivo-like conditions for systems biology. Febs J 277(3):749-
60.
van Eunen K, Bouwman J, Lindenbergh A, Westerhoff HV, Bakker BM. 2009. Time-
dependent regulation analysis dissects shifts between metabolic and gene-
expression regulation during nitrogen starvation in baker's yeast. Febs J
276(19):5521-36.
van Gulik WM. 2010. Fast sampling for quantitative microbial metabolomics. Curr Opin
Biotechnol 21(1):27-34.
van Maris AJ, Winkler AA, Kuyper M, de Laat WT, van Dijken JP, Pronk JT. 2007.
Development of efficient xylose fermentation in Saccharomyces cerevisiae: xylose
isomerase as a key component. Adv Biochem Eng Biotechnol 108:179-204.
Villas-Bôas SG. 2007a. Microbial metabolomics: Rapid sampling techniques to investigate
intracellular metabolite dynamics - an overview. In: Villas-Bôas SG, Roessner U,
Hansen MAE, Smedsgaard J, Nielsen J, editors. Metabolome analysis: An
introduction: John Wiley & Sons, Inc. p 203 - 214.
Villas-Bôas SG. 2007b. Sampling and sample preparation. In: Villas-Bôas SG, Roessner U,
Hansen MAE, Smedsgaard J, Nielsen J, editors. Metabolome analysis: An
introduction: John Wiley & Sons, Inc. p 39 - 82.
Villas-Bôas SG, Åkesson M, Nielsen J. 2005a. Biosynthesis of glyoxylate from glycine in
Saccharomyces cerevisiae. FEMS Yeast Res 5(8):703-9.
44 Metabolomics

Villas-Bôas SG, Bruheim P. 2007. Cold glycerol-saline: the promising quenching solution for
accurate intracellular metabolite analysis of microbial cells. Anal Biochem
370(1):87-97.
Villas-Bôas SG, Delicado DG, Åkesson M, Nielsen J. 2003. Simultaneous analysis of amino
and nonamino organic acids as methyl chloroformate derivatives using gas
chromatography-mass spectrometry. Anal Biochem 322(1):134-8.
Villas-Bôas SG, Hojer-Pedersen J, Åkesson M, Smedsgaard J, Nielsen J. 2005b. Global
metabolite analysis of yeast: evaluation of sample preparation methods. Yeast
22(14):1155-69.
Villas-Bôas SG, Moxley JF, Åkesson M, Stephanopoulos G, Nielsen J. 2005c. High-
throughput metabolic state analysis: the missing link in integrated functional
genomics of yeasts. Biochem J 388(Pt 2):669-77.
Visser D, Heijnen JJ. 2002. The mathematics of metabolic control analysis revisited. Metab
Eng 4(2):114-23.
Visser D, Heijnen JJ. 2003. Dynamic simulation and metabolic re-design of a branched
pathway using linlog kinetics. Metab Eng 5(3):164-76.
Visser W, Scheffers WA, Batenburg-van der Vegte WH, van Dijken JP. 1990. Oxygen
requirements of yeasts. Appl Environ Microbiol 56(12):3785-92.
Wang L, Birol I, Hatzimanikatis V. 2004. Metabolic control analysis under uncertainty:
framework development and case studies. Biophys J 87(6):3750-63.
Wisselink HW, Cipollina C, Oud B, Crimi B, Heijnen JJ, Pronk JT, van Maris AJ. 2010.
Metabolome, transcriptome and metabolic flux analysis of arabinose fermentation
by engineered Saccharomyces cerevisiae. Metab Eng 12(6):537-51.
Wisselink HW, Toirkens MJ, del Rosario Franco Berriel M, Winkler AA, van Dijken JP, Pronk
JT, van Maris AJ. 2007. Engineering of Saccharomyces cerevisiae for efficient anaerobic
alcoholic fermentation of L-arabinose. Appl Environ Microbiol 73(15):4881-91.
Wisselink HW, Toirkens MJ, Wu Q, Pronk JT, van Maris AJ. 2009. Novel evolutionary
engineering approach for accelerated utilization of glucose, xylose, and arabinose
mixtures by engineered Saccharomyces cerevisiae strains. Appl Environ Microbiol
75(4):907-14.
Wu L, Mashego MR, van Dam JC, Proell AM, Vinke JL, Ras C, van Winden WA, van Gulik
WM, Heijnen JJ. 2005. Quantitative analysis of the microbial metabolome by isotope
dilution mass spectrometry using uniformly 13C-labeled cell extracts as internal
standards. Anal Biochem 336(2):164-71.
Wu L, Wang W, van Winden WA, van Gulik WM, Heijnen JJ. 2004. A new framework for
the estimation of control parameters in metabolic pathways using lin-log kinetics.
Eur J Biochem 271(16):3348-59.
Yoshida S, Imoto J, Minato T, Oouchi R, Sugihara M, Imai T, Ishiguro T, Mizutani S, Tomita
M, Soga T and others. 2008. Development of bottom-fermenting Saccharomyces
strains that produce high SO2 levels, using integrated metabolome and
transcriptome analysis. Appl Environ Microbiol 74(9):2787-96.
Zaldivar J, Borges A, Johansson B, Smits HP, Villas-Boas SG, Nielsen J, Olsson L. 2002.
Fermentation performance and intracellular metabolite patterns in laboratory and
industrial xylose-fermenting Saccharomyces cerevisiae. Appl Microbiol Biotechnol
59(4-5):436-42.
Zamboni N, Kümmel A, Heinemann M. 2008. anNET: a tool for network-embedded
thermodynamic analysis of quantitative metabolome data. BMC Bioinformatics
9:199.
Part 2

Data Analysis and Integration


3

Online Metabolomics Databases and Pipelines


Adam J. Carroll
The Australian National University
Australia

1. Introduction
As metabolomics becomes an increasingly major component of modern biological research,
steps must be taken to preserve and make maximal use of the ever increasing torrents of
new data entering the public domain. While this task is by no means unique to the field of
metabolomics, the complexity, heterogeneity and large sizes of metabolomics datasets make
the development of effective metabolomics bioinformatics tools particularly challenging.
Despite these challenges, metabolomics specialists have recently been making rapid
progress in this area. A wide range of powerful web-based tools designed to facilitate the
systematic online storage, processing, dissemination and biological interpretation of
technically and biologically diverse metabolomics datasets have now emerged and are
rapidly becoming cornerstones of advancement in biological science.
Web-based tools for metabolomics perform a wide variety of functions. These can be
divided into several broad categories, including:
1. Storage and dissemination of technical, biological, and physicochemical reference data
for metabolites
2. Processing of raw instrument data to generate [metabolite x sample] data matrices
suitable for statistical and multivariate data-analysis
3. Database storage and querying of pre-processed relative and/or absolute metabolite
level data
4. Statistical and multivariate analysis of pre-processed relative and/or absolute
metabolite level data
5. Aiding biological interpretation of metabolomics results by integration of biological
knowledge such as known biomarkers or metabolic pathway information.
While some tools are broader in scope than others and some tools can essentially fully
service the data-processing requirements of certain metabolomics approaches, it is
important to note that no single tool is currently capable of fulfilling every requirement of
every metabolomics researcher. This chapter will review the current state of development in
the area of web-based informatics tools for metabolomics and explain how currently
available tools can be used to accelerate scientific discovery. It will then attempt to predict
future developments in the area of metabolomics web-tool development and advise new
metabolomics researchers on strategies to maximise their own benefit from these
developments.
48 Metabolomics

2. Information about metabolites: Biological cheminformatics


2.1 Background
One of the fundamental questions of metabolomics is “how many metabolites occur in
nature, what are their structures, what are their physical, chemical and biological properties
and how are they distributed amongst species?”. Large-scale efforts to build comprehensive
databases of metabolite-related knowledge are beginning to provide at least approximate
answers to these questions. Defining “the metabolome” of an organism in qualitative sense,
by building well-annotated catalogues of metabolites and their properties, is analogous to
sequencing and mapping the genome of an organism. That is, it provides a crucial
foundation for the development of analytical approaches and experiments, aids in the
interpretation of analytical results and provides an important scaffold upon which to attach
new information as it becomes available.
Because metabolites are small molecule chemicals of biological origin, organisation of
metabolite information lies at the interface between bioinformatics (the management of
biological information) and cheminformatics (the management of chemical information).
While metabolomics researchers will find useful information about metabolites in broad-
scoped, general cheminformatic databases, a new generation of biology-focused
cheminformatic databases are making it easier for biologists to find cheminformatic data
specifically related to biology. This section will guide the reader towards online sources of
metabolite information and explain how these information sources can be used to aid
metabolomics research.

2.2 Molecular semantics: The metabolite naming issue


One of the challenges associated with finding online information about metabolites can be
figuring out what text to enter into search engines. Metabolites can be named in many
different ways in many different places online and searching with one name will generally
only retrieve resources tagged with that particular name. Moreover, in cutting-edge
metabolomics research, it is frequently the case that one is searching for information about a
poorly-known or even completely hypothetical metabolite for which one has a structure in
mind but for which its common name, if indeed it has one, is unknown. Fortunately, there
are ways around these problems, thanks to the thoughtful design of cheminformatic
databases. These will be explained below.
For well-known metabolites, finding detailed information is particularly easy. Most
metabolite information databases annotate each metabolite entry with a large set of
‘synonyms’ – a range of different names commonly used to refer a given metabolite. As a
result, if one uses a common name to search those databases for information about a well-
known metabolite, one will usually find the information they need. In those cases, the key
thing is to know which databases to search (these will be outlined shortly).
Finding information on well-known metabolites is relatively easy. However, metabolomics
researchers are often interested in discovering new metabolites or learning what little is
known about more about poorly-known metabolites. Often, a researcher may know the
structure of a theoretical metabolite but have no idea whether it has been observed in nature
before let alone what its common name might be. Indeed, such ‘theoretical’ metabolites
Online Metabolomics Databases and Pipelines 49

often have been observed in nature before and have a common name, but finding this out
can be challenging if one does not know where to start. This is where InChI codes and
comprehensive InChI-enabled cheminformatic databases become indispensible (Wohlgemuth
et al., 2010).
“InChI” is an abbreviation for “International Chemical Identifier”, a system of expressing
chemical structures as compact strings of text suitable for efficiently and unambiguously
conveying chemical structures across text-based systems such as web search engines. The
InChI system was developed by the International Union of Pure and Applied Chemistry
(IUPAC) and the National Institute of Standards and Technology (NIST). Each unique
chemical structure can be converted into its own unique InChI code and vice-versa1. There
are a range of freely-available software tools that allow one to draw a chemical structure and
obtain its InChI code or enter an InChI code and have its structure drawn automatically (see
Table 1 for examples). All the major metabolite information databases tag their entries with
InChI codes, so if one is uncertain of the name of a target metabolite, the best approach is to
generate its InChI code and search with that. Some cheminformatic databases provide web-
based structure drawing tools allowing users to effectively generate an InChI code and
search with it in a single step. One of the advantages of using an unambiguous structural
identifier such as InChI to search a database is that if no hits are obtained, one may fairly
safely conclude that the target molecule was not in the database2. When a hit is obtained,
however, the returned information may include common name(s) for the molecule that can
aid in subsequent literature searches. For anyone building metabolite databases or
supplying supplementary tables of metabolite data for publication, annotation of these data
with InChI codes is highly-recommended (Wohlgemuth et al., 2010). Online tools for
generating InChI codes from structures or other identifiers are listed in Table 1. A
particularly useful tool for metabolomics researchers is the Chemical Translation Service
provided by the lab of Oliver Fiehn (Wohlgemuth et al., 2010) since this tool is capable of
batch translations of miscellaneous metabolite identifiers and synonyms to standard InChI
codes and other common identifiers.

2.3 Chemical ontologies: Organising metabolites into useful categories


In scientific communication, biologists frequently refer to broad ‘classes’ of metabolites
using terms related to their functional groups (eg. ‘alcohols’), their chemical properties (eg.
‘organic acids’) or biological roles (eg. ‘hormones’). Moreover, researchers are often
interested in obtaining lists of metabolites that fall in a particular class. For example, a
researcher may want to identify metabolites in an organism that contain a particular
functional group and will therefore be expected to undergo certain chemical reactions.
Potential classes range in scope from very broad (eg. ‘organic’) to moderately specific (eg.
‘alkaloids’) to even more specific (eg. ‘monoterpenoid indole alkaloids’) and so on. While
“metabolite classes” like these appear throughout the scientific literature, formalising them

1 There is one caveat to this statement. The only truly non-ambiguous InChI codes are called “Standard”

InChI (often abbreviated to “StdInChI” - these always begin with the string “InChI=1S/”). If building a
metabolomics database, it is advisable to use only standard InChI codes.
2 Some metabolite databases were built prior to the release of Standard InChI and have been annotated

using non-standard InChI codes (always beginning with “InChI=1/”). It is always a good idea to check
which InChI type a database uses before searching it with an InChI code.
50 Metabolomics

into accurately and systematically defined and ‘chemical ontologies’ that can be used in
practically useful ways is a non-trivial task. Despite this, a number of metabolite-related
databases have begun developing and/or employing hierarchical systems of compound
classification, allowing users to browse lists of metabolites via classification trees
(ontologies). Examples of databases employing compound ontologies or hierarchical
compound taxonomies for annotation of metabolite information include PubChem, ChEBI
(Degtyarenko et al., 2008), the BioCyc family of metabolic pathway databases (Caspi et al.,
2010), the Human Metabolome Database (HMDB) (Wishart et al., 2007) and
MetabolomeExpress (Carroll et al., 2010). The ChEBI compound ontology is by far the most
advanced and comprehensive ontology for biological small molecules and is downloadable
in open formats from the ChEBI website. Its adoption is recommended in the development
of new metabolomics databases.

Tool (URL) Features

Input: Structure drawing tool


ChEBI Advanced Search
Output: StdInChI, SMILES
(http://www.ebi.ac.uk/chebi/
Search capabilities: substructure,
advancedSearchForward.do#)
similarity, identical structure

Input: Structure drawing tool


Output: StdInChI, StdInChIKey, SMILES,
PubChem Structure Search
SMARTS, Formula
(http://pubchem.ncbi.nlm.nih.gov/
Search capabilities: substructure,
search/search.cgi)
superstructure, similarity, identical
structure

Input: Any major database ID, common


synonym or structure identifier
Fiehn Lab Chemical Translation Service Output: Any major database ID, common
(http://uranus.fiehnlab.ucdavis.edu:8080/ synonym or structure identifier
cts/homePage) Search capabilities: simple search by any
major database ID, common synonym or
structure identifier

Table 1. Recommended online tools for generating unambiguous InChI structural identifier
strings from structures, names or other identifiers

2.4 Physicochemical information


Physicochemical information about metabolites includes information about their physical
and chemical properties such as their structures, molecular formulas, molecular weights,
melting and boiling points, solubilities in different solvents at different temperatures,
polarities, pKa, light absorbance and fluorescence properties, energy contents, refractive
indices and other similar types of basic empirical information. This kind of information can
be extremely useful when designing extraction, sample clean-up or analyte-enrichment
protocols, for example.
Online Metabolomics Databases and Pipelines 51

2.5 Recommended sources of general metabolite information


Many online databases offer information about metabolites. These have varying scales and
scopes of content, query tools and modes of access. In these aspects, several databases stand
out from all others and these are described below.

2.5.1 ChemSpider
Description: A freely-accessible collection of compound data from across the web with a
very versatile search engine.
Scope: all chemicals – not just metabolites
Semantic content: Many synonyms, identifiers and external database IDs and link-outs
Physicochemical content: Masses, formula, experimental melting point, physical state,
appearance, stability, storage compatibility, safety. A substantial amount of additional
predicted data.
Biological content: Links to MeSH
Analytical content: Some compounds have spectra
Noteworthy tools: Search by physicochemical properties
Modes of access: search (by synonym, InChI, SMILES, CAS, structure), API. Limited to 5000
structures per day.
Strengths: Enormous index of chemicals that is widely linked to external online resources –
a good starting point if looking for information on a particular chemical.
Limitations: Broad focus means extracting desired subsets of information can be difficult.
Cannot be downloaded. Results only returned in HTML - not spread sheet format. Limited
to 5000 structures per day.
URL: http://www.chemspider.com/

2.5.2 Human Metabolome Database (HMDB)


Description: A comprehensive, freely-available knowledgebase of human metabolite
information.
Scope: Human (Homo sapiens) metabolites
Semantic content: metabolite names, formulas, masses, structures, InChI, SMILES, external
database IDs and link-outs; Chemical taxonomy
Physicochemical content: Masses, formula, water solubility, hydrophobicity, melting point,
physiological charge, physical state
Biological content: Presence in human cellular compartments and biofluids; Measured
concentrations in biofluids; reactions; enzymes; enzyme genes; disease associations;
descriptions of biological roles
52 Metabolomics

Analytical content: many compounds have LC/MS, GC/MS and/or NMR spectra obtained
under standardised conditions
Noteworthy tools: Versatile ‘Data Extractor’. Searching based on spectral properties
Modes of access: browse, search, complex query with data extractor, download
Strengths: Comprehensive, may be freely downloaded in entirety. Human focus is good for
human metabolomics researchers.
Limitations: Important fields are empty for some very common metabolites. Being limited
to human metabolites limits utility for other research areas. Downloadable flat-file format
requires parsing in order to be usable in spread sheets or local databases.
Reference: (Wishart et al., 2007)
URL: http://www.hmdb.ca/

2.5.3 Chemical Entities of Biological Interest (ChEBI)


Description: A freely-available dictionary of small molecule chemicals of interest to
biologists.
Scope: Small molecules of biological interest (endogenous biochemicals and exogenous
bioactive compounds)
Semantic content: metabolite names, formulas, masses, structures, InChI, SMILES, external
database IDs and link-outs; ChEBI Chemical Ontology
Physicochemical content: Formal charge
Biological content: None
Analytical content: None
Noteworthy tools: Structure based search, Versatile advanced query, Chemical and
functional ontology-based browsing
Modes of access: browse, search, complex query, FTP download
Strengths: May be downloaded in bulk. Versatile advanced query tool. Query results
downloadable in useful formats. Well-designed ontology.
Limitations: Human-centric. Far from comprehensive, particularly for non-human-related
information. References to supporting literature are not provided with biofunctional
ontology assignments. No species occurrence information.
Reference: (Degtyarenko et al., 2008)
URL: http://www.ebi.ac.uk/chebi

2.5.4 PubChem
Description: A freely available general dictionary of chemicals.
Online Metabolomics Databases and Pipelines 53

Scope: Any small molecules


Semantic content: metabolite names, formulas, masses, structures, InChI, SMILES, external
database IDs and link-outs; MeSH chemical classification
Physicochemical content: Formal charge, partition coefficient, H-bonding donor and
acceptor counts
Biological content: Bioactivity, bioassay results, safety and toxicology, associations with
metabolic pathways in KEGG
Analytical content: None
Noteworthy tools: Structure and structural similarity-based search, versatile advanced
query, chemical and biomedical ontology-based browsing, chemical structure clustering
Modes of access: browse, search, complex query, FTP download
Strengths: Huge number of compounds. Highly annotated. Some information is available
for bulk download. Versatile advanced query tool. Extensive link-outs.
Limitations: Much compound information displayed on the website is not provided for
bulk download. No species occurrence information.
URL: http://pubchem.ncbi.nlm.nih.gov

2.6 Metabolic pathway databases


A wide range of biological information about metabolites is available online. Utilising
this information can aid in the development of hypotheses, the design of experiments
and the biological interpretation of metabolomics results. For this purpose, among the
most useful types of database are metabolic pathway databases. These play a crucial role
in metabolomics research by systematically capturing and providing a close
representation of current knowledge about: a) which metabolites occur in particular
biological systems; b) the enzymatic and non-enzymatic reactions that link different
metabolites together into metabolic pathways; c) the enzymes that carry out these
reactions and the genes that encode them; and d) the allosteric interactions and
signalling networks that regulate these genes and gene products. Another highly useful
function that some metabolic pathway databases carry out is to visually overlay
metabolomic datasets over pathway diagrams to provide biological contexts aiding the
biological interpretation of results. Some of most useful metabolic pathway databases are
described below.

2.6.1 Kyoto Encyclopedia of Genes and Genomes (KEGG)


Description: A knowledgebase of genomes, genes, gene-products their properties and the
metabolic and regulatory pathways they form.
Species: many species from many different classes
Metabolic pathway content: metabolite names, formulas, masses, structures and external
database IDs; reactions; reactant-product atom mappings; pathways; enzymes; enzyme
54 Metabolomics

genes; orthologies; bioactivities; allosteric interactions / regulatory pathways; pathway,


compound, taxonomy and biological process ontologies
Noteworthy features: Structural similarity search
Modes of access: browse, search, API, FTP download (requires subscription)
Strengths: Enormous amount of information. The largest source of atom-mapped reactions
available.
Limitations: Broad focus means extracting desired subsets of information can be
challenging. Query tools are limited.
Reference: (Ogata et al., 1999)
URL: http://www.genome.jp/kegg/

2.6.2 BioCyc and the “Cyc“ family of metabolic pathway databases


Description: Similar to KEGG. A collection of Pathway / Genome Databases (PGDBs) built
using software that predicts metabolic pathways from genome sequences and subsequently
refined by varying degrees of expert curation.
Species: BioCyc itself includes highly-curated PGDBs for 3 organisms: Escherichia coli
(EcoCyc), Arabidopsis thaliana (AraCyc), Saccharomyces cerevisiae (YeastCyc). Another highly-
curated PGDB called MetaCyc compiles pathway and enzyme information from >1900
organisms (mainly single-cell organisms) into a single reference database. See also the
separate HumanCyc, PlantCyc and many other “Cyc” databases.
Metabolic pathway content: metabolite names, formulas, masses, structures, InChI,
SMILES, Gibbs free energies and external database IDs; reactions; pathways; enzymes;
enzyme genes; allosteric interactions / regulatory pathways; compound, pathway, gene and
enzyme ontologies; links to literature supporting pathways
Noteworthy features: Interactive cellular overview pathway display, Regulatory overview,
Genome browser, Advanced query tool, Powerful API, Omics viewer
Modes of access: browse, search, API, FTP
Strengths: Enormous amount of information. Powerful and intuitive query tools and API
make extraction of data subsets easy. Returns results in simple tables and open XML
pathway exchange formats. Omics viewer allows overlaying of omics (including
metabolomics) data onto pathway views.
Limitations: Some useful and easily-fillable fields are empty for some metabolites. The
Cyc databases often refer to generic entities such as ‘a fatty acid’ – this can limit their
utility when researchers are interested in modelling connections between certain specific
entities.
Reference: (Caspi et al., 2010)
URLs: http://biocyc.org, http://humancyc.org, http://www.plantcyc.org and others
Online Metabolomics Databases and Pipelines 55

2.6.3 Reactome
Description: An interactive collection of curated, peer-reviewed metabolic pathways with
cross-referencing of reactions and pathways between organisms. Pathways are displayed
via an intuitive GUI but may be downloaded in a variety of open formats.
Species: A variety of species. Most comprehensive for human.
Metabolic pathway content: hierarchically organised curated and peer-reviewed metabolic
pathways; reactions; reaction-gene associations
Noteworthy features: Interactive pathway viewer
Modes of access: browse, search and download
Strengths: Peer-reviewed, user-friendly, different subcellular metabolite pools are treated as
separate entities
Limitations: Reaction-centric. Not much information about metabolites and does not
provide any tools for overlaying metabolite expression data.
Reference: (Croft et al., 2011)
URL: http://www.reactome.org

2.6.4 KappaView
Description: A web-based tool allowing users to overlay metabolite- and gene-expression
responses and correlations onto custom pathway diagrams or onto a collection of neat,
simple and interactive metabolic pathway diagrams.
Species: A variety of species.
Metabolism-related content: hierarchically organised curated metabolic pathways;
reactions; reaction-gene associations
Noteworthy features: Gene and metabolite expression overlay
Modes of access: browse, search and download
Strengths: User-friendly; neat/simple diagrams; may be integrated into third party websites
using a flexible API; can also overlay metabolite-metabolite, gene-gene and metabolite-gene
correlations
Limitations: Does not support InChI
Reference: (Sakurai et al., 2010)
URL: http://kpv.kazusa.or.jp/kpv4/

2.6.5 Human Metabolome Database (HMDB) Pathways


Description: A comprehensive, freely-available knowledgebase of human metabolite
information.
56 Metabolomics

Species: Human (Homo sapiens)


Metabolism-related content: A set of ‘textbook’ style metabolic pathway diagrams with
metabolites hyperlinked to HMDB metabolite information pages and enzymes hyperlinked
to UniProt database.
Noteworthy features: None
Modes of access: browse, search
Strengths: Easy to understand
Limitations: Not downloadable. No documented API.
Reference: (Wishart et al., 2007)
URL: http://www.hmdb.ca

2.6.6 KNApSAcK
Description: A comprehensive species-metabolite relationship database for plants.
Although not strictly a metabolic pathway database, this database is useful for identifying
plant species that contain a certain chemical or identifying chemicals that have been
reported in a particular plant species or higher level taxon.
Species: Plants
Metabolism-related content: References to literature reporting the presence of compounds
in different plant species. Chemical structures. Masses.
Noteworthy features: References to literature.
Modes of access: browse, search
Strengths: Contains information on many plant-specific specialised metabolites.
Limitations: Data itself is not downloadable.
Reference: (Shinbo et al., 2006)
URL: http://kanaya.naist.jp/KNApSAcK/

3. Online analytical reference spectra for metabolomics


3.1 The roles of analytical reference libraries in metabolomics research
The first online metabolomics databases to store and disseminate actual instrument data
for metabolites generally provided spectral reference libraries. These spectral libraries
provide reference signals for authentic standard compounds and sometimes also for
‘unknown’ metabolites obtained through the analysis of standards and biological
materials under controlled conditions. The de-novo construction of large analytical
reference libraries requires expertise in chemistry, is time consuming and expensive.
Centralization of spectral reference data in expert-curated public repositories helps the
metabolomics community by: 1) making it easier and cheaper for new labs to build their
Online Metabolomics Databases and Pipelines 57

own data processing pipelines; 2) reducing the probability of metabolite misidentification


by non-specialists; and 3) promoting efficient communication about ‘unknown’
metabolites that are recognisable on the basis of their analytical properties but for which
no structural information is available.

3.2 Types of analytical reference spectra available online


Reference spectra are available from a number of online sources. Types of reference data
available include downloadable mass-spectral and retention-index (MSRI) libraries for gas
chromatography / mass spectrometry (GC/MS) (Kopka et al., 2005; Schauer et al., 2005;
Carroll et al., 2010), searchable but not-downloadable MSRI data (Skogerson et al., 2011),
NMR spectra collected under standardized conditions (Wishart et al., 2007; Cui et al.,
2008; Ulrich et al., 2008) and MS and MS/MS spectra from a wide range of platforms
including accurate mass instruments (Smith et al., 2005; Horai et al., 2010). In addition,
most cheminformatic and metabolic pathway databases provide accurate monoisotopic
mass information for metabolites which can help provide candidate identities for
accurate-mass LC/MS and direct-infusion (DI)/MS peaks. These data sources are
described in detail later.

3.3 Reference data for Nuclear Magnetic Resonance (NMR)


One of the great advantages that NMR has over mass-spectrometry is that chemical shifts
and coupling constants – unlike mass-spectral fragmentation patterns - are, under readily
controllable conditions, absolute physical constants that may be readily and accurately
reproduced between different makes and model of instrument. Reference libraries of NMR
spectra of metabolites, acquired under standardized conditions, are therefore of broad
utility by the metabolomics research community. The major sources of standardized NMR
spectra for metabolomics are the Madison Metabolomics Consortium Database (Cui et al.,
2008), the Biological Magnetic Resonance Bank (Ulrich et al., 2008) and the HMDB (Wishart
et al., 2007). These are detailed shortly.

3.4 Reference data for Gas-Chromatography / Mass-Spectrometry (GC/MS)


The most useful reference data for GC/MS are downloadable MSRI libraries. These are
libraries of mass-spectra and retention indices for peaks observed in GC/MS
chromatograms obtained by the GC/MS analysis of pure compounds and biological
samples under standardised conditions (Kopka et al., 2005; Schauer et al., 2005). When the
same standardized conditions are employed for GC/MS analysis in different laboratories, a
single common MSRI library can be used for the high-confidence identification of common
metabolite signals in those different labs (Schauer et al., 2005). Researchers setting up new
GC/MS metabolomics platforms are advised to consider adopting a standardised GC/MS
protocol already supported by a publicly-available MSRI library such as those available
from the Golm Metabolome Database (Kopka et al., 2005) or MetabolomeExpress (Carroll et
al., 2010) since this will enable them to share MSRI libraries with those labs and benefit from
ongoing efforts to extend those libraries and annotate the large number of ‘unknown’
metabolites detected in GC/MS chromatograms of biological samples.
58 Metabolomics

n
3.5 Reference data for liquid chromatography-MS, MS/MS and MS
While the low-cost and operational simplicity of GC/MS has led it to become the most
widely employed analytical platform in metabolomics, an increasing number of laboratories
are adopting complementary techniques based on liquid chromatography (LC)- and direct
infusion (DI)/MS methods that employ different ionisation techniques and more advanced
mass-spectrometers capable of MS, MS/MS, MS3 and MSn modes of analysis together with
much higher mass accuracy and resolution than is provided by most standard GC-MS
systems. In the paragraphs below, the various types of non GC/MS, MS-based
metabolomics techniques such as LC/MS, DI/MS and capillary electrophoresis (CE)/MS
including tandem MS and MSn methods will be referred to collectively as “LC/MS”
techniques.
While GC/MS metabolomics is dominated almost entirely by electron impact ionisation (EI)
methods using the industry-standardised ionisation energy of 70eV, yielding highly-
reproducible fragmentation spectra between different GC/MS instruments, such broad
standardisation has not occurred for LC/MS. For LC/MS, the enormous diversity of mass-
spectrometer types, combined with a lack of highly-developed LC ‘retention-index’ systems
present significant challenges towards the creation of standardized MSRI reference libraries,
analogous to those available for GC/MS, capable of unambiguous cross-laboratory peak
identification for LC/MS.
The simplest type of online reference data for LC/MS metabolomics are the accurate,
monoisotopic masses and molecular formulas of metabolites and, in some cases, their stable-
isotope-labelled isotopomers. The data-processing packages provided with MS instruments
capable of high-accuracy mass measurements generally allow users to create custom
libraries of accurate masses and/or molecular formulas (for improved match scoring based
on the shapes of isotopic envelopes) for target analytes to assist with peak identification.
Although accurate masses or molecular formulas alone are not sufficient to unambiguously
identify metabolite signals (due to the high frequency of structural isomers across nature),
using these data in a rational manner can often provide valuable clues about the possible
identities of peaks.
A good way of reducing (but not eliminating) ambiguity in accurate mass-based assignments
is to build a separate accurate mass library for each biological system under investigation
and to include in each library only those metabolites for which literature evidence exists to
support their presence in that organism. An easy way of doing this is to use the advanced
query tool provided with each of the BioCyc family of metabolic pathway databases (of
which there are many). While the metabolite sets thus obtained may not be complete, this is
a fast way of obtaining a good quality starting set.
Another approach for reducing ambiguity in LC/MS peak identifications is to use MS/MS
spectral similarity as a scoring parameter to complement accurate-mass MS based
assignments (see (Matsuda et al., 2009; Matsuda et al., 2010) for good examples). The major
online sources of MS/MS spectra for metabolites are MassBank (Horai et al., 2010), METLIN
(Smith et al., 2005), ReSpect for Phytochemicals (http://spectra.psc.riken.jp/menta.cgi/
index) and the HMDB (Wishart et al., 2007). These databases each have different strengths
and limitations which will be outlined shortly. With the notable exception of ReSpect for
Phytochemicals, a drawback that these databases share is a lack of support for bulk
Online Metabolomics Databases and Pipelines 59

downloading of spectra. That said, MassBank does provide a powerful API to partially
overcome the need for bulk download while the METLIN website currently reports that an
API is in development.

3.6 The need for chromatographic retention data in LC/MS reference databases
It is important to note that, for high-confidence peak identifications that meet minimum
reporting standards outlined by the Metabolomics Standards Initiative (MSI) (Sansone et al.,
2007), it is necessary to support peak identifications with an additional, orthogonal
identification parameter. In the case of LC/MS, where chromatography is used, this
parameter is generally retention time or relative retention time agreement with an authentic
standard. Unfortunately, there appear to be few if any LC/MS reference databases that
provide retention time or relative retention time information. Absolute retention times vary
from instrument to instrument and from column to column (even between columns of the
same make and model), and are therefore considered to be of limited use for high-
confidence inter-laboratory peak identification. However, relative retention times (or
retention indices), where the retention time of each peak is expressed relative to one or two
other peaks in the same chromatogram, are far more stable (Tarasova et al., 2009) and may
provide an avenue to the compilation of LC-MS reference libraries capable of providing
MSI-compliant peak identifications by combining accurate mass MS or MS/MS spectra with
meaningful and highly reproducible retention index (RI) properties. Complementary to this
approach would be the further development of RI-prediction models that can accurately
predict the LC retention indices of metabolites based on their structures (Hagiwara et al.,
2010).
It is important to note that sufficient RI reproducibility may only be achievable with certain
simple types of stationary and mobile phase combinations whereby a single stationary
phase interaction mechanism (eg. hydrophobic interactions in C18 reversed-phase
chromatography or hydrogen-bonding interactions in silanol based normal phase
chromatography) applies to all analytes. In separations over mixed-mode stationary phases
where multiple interaction mechanisms occur, there is more potential for variations in
chromatographic conditions to differentially affect different peaks, thus changing their
relative retention times. Public databases of “Accurate Mass / retention Time (AMT) tags”
are playing increasingly important roles in peptide identification in LC-MS proteomics
(Hagiwara et al., 2010). A similar trend is to be expected in metabolomics.

3.7 Major online sources of analytical reference spectra for metabolomics


3.7.1 Madison Metabolomics Consortium Database (MMCD)
Description: An analytical reference database and signal-matching tool for metabolomics.
Species: Not species-constrained
Reference data: Standardized NMR spectra for 791 different metabolites (1H, 13C, DEPT90,
DEPT135, [1H, 1H] TOCSY and [1H, 13C] HSQC). General information on >20 000
metabolites.
Noteworthy features: NMR spectrum-based search. Batch search capability.
60 Metabolomics

Modes of access: browse, search and download individual spectra via web interface. Bulk
FTP download of raw spectra via BMRB FTP site.
Strengths: Enormous resource for NMR metabolomics. Includes a wide range of metabolites
including those that don’t occur in humans (eg. plant-specific metabolites). Spectral
matching tools provide batch-processing capability.
Limitations: No support for bulk download of metabolite information based on complex
query
Reference: (Cui et al., 2008)
URL: http://mmcd.nmrfam.wisc.edu

3.7.2 Human Metabolome Database (HMDB)


Description: A comprehensive, freely-available knowledgebase of human metabolite
information.
Species: Human (Homo sapiens)
Reference data: Standardized MS/MS and NMR spectra (1H, 13C, 13C HSQC, TOCSY) for
>780 metabolites. GC/MS MSRI reference data on ~300 metabolites.
Noteworthy features: NMR, MS/MS and GC/MS spectrum-based search
Modes of access: browse, search and bulk download (bulk download of MS/MS spectra
only provides images of spectra).
Strengths: A large set of standardized NMR and GC/MS spectra help new labs to quickly
set up metabolite profiling platforms.
Limitations: No support for bulk download of metabolite information based on complex
query. No batch-processing capabilities for spectral matching. No API for integration with
other web tools.
Reference: (Wishart et al., 2007)
URL: http://www.hmdb.ca

3.7.3 METLIN
Description: A repository for metabolite information and tandem mass spectrometry data.
Species: Not formally species-constrained but is fairly human-centric
Reference data: Accurate masses of >44000 metabolites. >28000 high-resolution
Quadrupole/Time-Of-Flight (Q/TOF) MS/MS spectra for ~5000 metabolites. Multiple
collision energies.
Noteworthy features: Batch searching of mzXML MS/MS files against the database.
Integration with XCMS LC/MS data-processing pipeline. Neutral loss search.
Modes of access: Search only. API in development
Online Metabolomics Databases and Pipelines 61

Strengths: A large set of standardized NMR and GC/MS spectra help new labs to quickly
set up metabolite profiling platforms.
Limitations: No bulk-download (must be purchased from instrument manufacturer).
Reference: (Smith et al., 2005)
URL: http://metlin.scripps.edu

3.7.4 MassBank
Description: A repository for mass-spectra of pure compounds. Features a unique design
involving a centralised interface but a distributed network of data servers providing the
mass-spectra.
Species: Not species constrained. Not limited to biological metabolites.
Reference data: >29000 mass spectra from a wide range of instrument types including, but
not limited to, GC/MS, LC/MS and LC-MS/MS.
Noteworthy features: Batch searching of MS/MS files against the database. Neutral loss
search. Most sophisticated and powerful spectral search and visualisation capabilities of all
available mass-spectral repositories.
Modes of access: Search, browse and API.
Strengths: Many spectra, powerful search capabilities.
Limitations: No bulk-download. However, individual spectra may be downloaded in text
format.
Reference: (Horai et al., 2010)
URL: http://www.massbank.jp/

3.7.5 ReSpect for Phytochemicals


Description: An interactive collection of MSn spectra of plant metabolites, collected by the
LC/MS metabolomics group of the RIKEN Plant Science Center.
Species: Plant species.
Reference data: A total of >8500 MS/MS spectra including >3000 spectra from the literature,
>4000 triple quadrupole MS/MS spectra corresponding to >861 standard compounds and
>1000 Q/TOF spectra corresponding to >550 standard compounds. Includes both +ve and –
ve ionization modes.
Noteworthy features: Spectral search online using cosine method
Modes of access: Search, browse and complete download.
Strengths: Contains many plant-specific spectra not available elsewhere. Free for bulk
download.
Limitations: No API. No batch search capability.
URL: http://spectra.psc.riken.jp/menta.cgi/index
62 Metabolomics

3.7.6 MS-MS Fragment Viewer 1.0


Description: A database of Liquid Chromatography Fourier Transfer Ion Cyclotron
Resonance Mass Spectroscopy (LC-FT/ICR-MS), Ion-Trap Tandem Mass Spectroscopy
(IT-MS/MS), Fourier Transform Tandem Mass Spectroscopy (FT-MS/MS) and
photodiode array (PDA) spectra with predicted structures of fragment ions observed in
LC-FT/ICR-MS.
Species: Plant species.
Reference data: Spectral data for 116 different flavonoids.
Noteworthy features: Predicted structures of fragment ions observed in LC-FT/ICR-MS.
Modes of access: Search only.
Strengths: Ultra-high mass accuracy of FT/ICR-MS.
Limitations: Limited range of spectra. Search only. No browse capability means it is
impossible to know what to search for. No spectral-based searching.
URL: http://webs2.kazusa.or.jp/msmsfragmentviewer/

3.7.7 MoTo DB
Description: A liquid chromatography-mass spectrometry-based metabolome database for
tomato
Species: Tomato (Solanum lycopersicum)
Reference data: Masses, retention times, UV/Vis properties and MS/MS fragment
information for a range of metabolites reported to occur in tomato plants.
Noteworthy features: Includes retention times.
Modes of access: Search only.
Strengths: Provides literature references to support peak annotations.
Limitations: Very limited search capability. No browse capability. No download.
Reference: (Moco et al., 2006)
URL: http://appliedbioinformatics.wur.nl/moto/

3.7.8 The Golm Metabolome Database (GMD)


Description: An interactive and downloadable database of electron impact (EI) ionization
mass-spectra and associated retention indices of metabolite peaks detected by GC-EI-
Quadrupole (GC-EI-Q-MS) and GC-EI-Time Of Flight (GC-EI-TOF-MS) instruments
operated under standardized conditions.
Species: Not formally species-constrained but is plant-centric.
Reference data: Contains MSRI data for ~4500 analytes (different chemical derivatives)
corresponding to ~1500 metabolites
Online Metabolomics Databases and Pipelines 63

Noteworthy features: Decision-tree based substructure prediction.


Modes of access: Search, browse and API
Strengths: Very comprehensive. Free for download. Well curated and supported.
Limitations: Does not provide innate support for sharing of MSRI libraries by arbitrary
users.
Reference: (Kopka et al., 2005)
URL: http://gmd.mpimp-golm.mpg.de/Default.aspx

3.7.9 MetabolomeExpress
Description: An interactive database of downloadable MSRI libraries, raw and processed
GC/MS metabolite profiling datasets and a database of metabolic phenotypes observed in
any organism using any analytical technique. Includes a complete GC/MS data processing
pipeline and cross-study data mining tools.
Species: Not formally species-constrained but current content is plant-centric.
Reference data: A number of GC/MS MSRI libraries are downloadable from the website. Golm
Metabolome Database MSRI libraries are provided for use within the data processing pipeline.
Noteworthy features: Members may independently upload their own MSRI libraries for
interactive dissemination and use within the GC/MS data-processing pipeline.
Modes of access: browse and FTP
Strengths: Libraries free for download. Provides a built-in GC/MS data processing
pipeline.
Limitations: No API. No search.
Reference: (Carroll et al., 2010)
URL: http://www.metabolome-express.org

4. Web-based data processing pipelines for metabolomics


4.1 Background
Less than a decade ago, software packages enabling processing and analysis of metabolomics
datasets were restricted to a limited range of desktop software programs. Would-be
metabolomics researchers would have to download or purchase and install software on local
computers, set up local reference libraries for peak identification and sometimes develop
custom in-house computer scripts to adapt the outputs of various programs into the formats
required by programs used for downstream analysis. These challenges were compounded by
the fact that available programs often lacked the kinds of specialised, biology-related features
desirable for metabolomics research. However, the understandable widespread dissatisfaction
of metabolomics researchers with this situation has, over the last decade, driven rapid
development of powerful online, platform-independent data processing pipelines tailored
64 Metabolomics

towards the needs of metabolomics research. Thanks to the availability of these packages and
the availability of standardised analytical reference libraries, it is now quite feasible for
researchers with limited experience to conduct detailed processing and analysis of their
instrumental datasets with little more than a fast internet connection, an up-to-date web-
browser and, in some cases, an FTP-client program for uploading data. This section will
provide an overview of the types of data processing pipelines that are currently accessible
online and compare the most powerful examples in more detail.

4.2 Functions carried out by online data-processing tools


Any ideal metabolomics data-processing pipeline, whether online or offline, should be able to:
a) identify and quantify biologically-relevant signals from raw instrument files and distinguish
them from biologically irrelevant signals; b) identify non-redundant metabolite signals and,
where possible, annotate them with their molecular identities; c) assemble a [metabolite x
sample] data matrix appropriately normalised to sample volumes, internal standards and/or
other useful normalisation factors; d) facilitate determination and statistical analysis of relative
metabolite levels between sample classes; e) carry out multivariate analyses such as principal
components analysis (PCA), hierarchical clustering analysis (HCA) and partial least squares
discriminant analysis (PLS-DA); and f) provide facilities to assist biological interpretation of
results (eg. mapping of detected metabolite responses onto metabolic pathways, over-
representation analysis and biomarker detection). While the vast majority of online
metabolomics data-processing tools carry out only one or a few of these functions, there are
systems capable of carrying out all of these functions. The functionalities of a variety of web-
based data processing tools for metabolomics are summarised in Table 2.

5. Online metabolomics data repositories


5.1 Background
The long-standing scientific tradition of openly disclosing supporting primary data whenever
scientific claims are made has been a fundamental factor underlying the credibility of science.
However, in more recent years, the scale and complexity of primary datasets has risen
dramatically, presenting ever-new challenges to this tradition with the widespread emergence
of high-throughput metabolomics technologies in bioscience being a good example.
In this author’s view, it is absolutely crucial that the culture of open primary data disclosure
is maintained, and that “challenges” should not become “excuses”. Even in the extreme case
of next-generation DNA sequencing where the sizes of typical primary datasets (after
parsing of raw image data) are typically measured in the 10’s of gigabytes (at least 10 times
larger than typical metabolomics datasets), scientists have risen to the challenge by
providing online storage space and developing specialised data repository systems capable
of systematically archiving and effectively disseminating these data (Kaminuma et al., 2010;
Cochrane et al., 2011; Leinonen et al., 2011).
Given the relatively small sizes of metabolomics datasets and the fact that metabolomics
techniques predate next-generation sequencing by a considerable number of years, it is
difficult to think of a satisfactory justification for the number of scientific claims that have
been made on the basis of metabolomics datasets that have not, at the very least, been made
freely available for download from a publicly accessible web site. That said, recent years
Online Metabolomics Databases and Pipelines 65

Feature / Tool Metabolome XCMS Metabo metDAT


MeltDB
Name: Express Online Analyst 2.0
(Neuweg
(Carroll et al., (Smith et (Xia et al., (Biswas et
Reference: er et al.,
2010) al., 2006) 2009) al., 2010)
2008)
GC/MS,
Raw data GC/MS,
GC/MS LC/MS LC/MS, DI/MS
processing LC/MS
NMR
Raw data
Interactive Static Static Static
visualization
Peak detection + + + + binning
accurate accurate
Peak ID method MSRI MS
mass mass
Ambiguou Ambiguo Ambiguo
Peak ID ambiguity Unambiguous
s us us
identification- COW COW
Peak alignment COW +
based (XCMS) (XCMS)
Processed data handling
Data matrix
+ + + + +
construction
Normalisation + + + +
Fold change
+ + + + +
calculation
Univariate t-test,
t-test t-test + +
statistics ANOVA
Correlation
+ + + +
analysis
Multivariate analysis
PCA + + + +
PLS-DA + + +
Cluster analysis + + + +
Classification and
+ +
feature analysis
Biological interpretation
Pathway mapping In development + +
Phenotype
+
recognition
ORA In development +
Table 2. Comparison of features of major web-based data processing pipelines for
metabolomics. Only tools capable of some level of raw data processing have been included.
A ‘+’ indicates the presence of a feature. COW = Correlation Optimized Warping (the
algorithm used by XCMS); MSRI = Mass Spectral and Retention Index; PCA = Principal
Components Analysis; PLS-DA=Partial Least Squares-Discriminant Analysis; ORA=Over-
representation Analysis; DI/MS = Direct Infusion / Mass Spectrometry
66 Metabolomics

have seen a strong increase in the number of metabolomics labs sharing primary datasets
from their own websites and even the emergence of centralized metabolomics data
repositories allowing arbitrary labs to share their datasets publicly without even having to
set up their own website. These groups that have been voluntarily driving the free and open
dissemination of primary metabolomics data should be commended! The following sections
will highlight the data sharing efforts that have been made by individual groups within the
metabolomics community and describe the centralized metabolomics data repositories that
are currently in operation and/or development.

5.2 Online databases sharing raw and/or partially-processed experimental datasets


5.2.1 DROP met: Data resources of plant metabolomics
Description: A part of the PRIMe (Platform for RIKEN Metabolomics) website.
Species: Plant species
Reference data: Provides a simple download page allowing free download of raw and/or
processed LC/MS and GC/MS datasets and metadata from 8 different peer-reviewed
publications emerging from the RIKEN Plant Science Center.
Noteworthy features: Metadata for each raw data file is provided in a systematic, MSI-
compliant format.
Modes of access: browse
Strengths: Data are easy to find and well annotated.
Limitations: Metabolic phenotypes are not stored in a database. There is no way of querying
the data without downloading, extracting biological information and importing into a local
database.
URL: http://prime.psc.riken.jp/?action=drop_index

5.2.2 KomicMarket (Kazusa omics data market)


Description: A freely accessible database of annotations of metabolite peaks from FT-ICR-
MS analysis of standard compounds and plant samples.
Species: Plant species
Reference data: Metabolites detected in tomato fruits by FT-ICR-MS. 215 standard
compounds detected by FT-ICR-MS.
Noteworthy features: None
Modes of access: Search, browse and API.
Strengths: Good collection of high mass-accuracy flavonoid spectra. API makes download
of spectra and associated annotations relatively easy.
Limitations: No bulk download of spectra needlessly makes access to spectra more
challenging.
Online Metabolomics Databases and Pipelines 67

Reference: (Iijima et al., 2008)


URL: http://webs2.kazusa.or.jp/komics/index.php

5.2.3 MassBase 1.0


Description: A mass-spectral tag archive for metabolomics.
Species: Plant species
Reference data: Provides raw and processed GC/MS, LC/MS and CE/MS data for
download.
Noteworthy features: None
Modes of access: Search, browse and download.
Strengths: One of very few sites to archive and disseminate raw chromatograms.
Limitations: No bulk-download of data – data files must be downloaded one at a time via
the web interface. Chromatograms provided in proprietary binary formats. Limited
metadata provided. Peak annotations are not provided.
URL: http://webs2.kazusa.or.jp/massbase/index.php/

5.2.4 SetupX
Description: A study design database for GC/MS metabolomics experiments.
Species: Plant species
Reference data: Provides raw and processed GC/MS data for download together with
metadata.
Noteworthy features: Metabolite detections are searchable by species and species are
searchable by metabolite detections.
Modes of access: Search, browse and download.
Strengths: One of very few sites to archive and disseminate raw chromatograms.
Experimental datasets may be downloaded as single zipped files.
Limitations: Enormous sizes of zipped experimental dataset files means that download
errors frequently occur during long downloads. No quantitative information is provided
with metabolite detections and there is no way to compare the results of different
experiments.
Reference: (Scholz and Fiehn, 2007)
URL: http://fiehnlab.ucdavis.edu:8080/m1/

5.2.5 PlantMetabolomics.org
Description: A database of processed, large-scale metabolic phenotype information
obtained from an array of different Arabidopsis thaliana T-DNA insertion mutants.
68 Metabolomics

Species: Arabidopsis thaliana


Reference data: Provides relative metabolite levels of a large number of metabolites in a
large number of Arabidosis thaliana mutants.
Noteworthy features: None.
Modes of access: Search, browse and download.
Strengths: Data on a very wide range of metabolites. Incorporates phenotypic notes on
mutants.
Limitations: Important metadata fields are frequently left empty. Raw data files are not
provided. Origins of processed results are not transparent. There is no way to align and
compare global phenotypes of mutants.
Reference: (Bais et al., 2010)
URL: http://plantmetabolomics.vrac.iastate.edu/ver2/index.php

5.2.6 Mery-B
Description: A repository for plant metabolomics datasets including experimental metadata
processed data and raw data for NMR experiments.
Species: Plants.
Reference data: Provides NMR-based metabolite quantification data for a variety of tissues
from a variety of species grown under a variety of conditions. Based on ~1000 spectra.
Chemical shift peak assignment information is provided.
Noteworthy features: Interactive raw data viewers for 1D NMR and GC/MS data.
Modes of access: Search and browse.
Strengths: Contains data from a range of peer-reviewed publications and references to
literature are clearly presented. Raw NMR spectra and GC chromatograms are available for
visualisation. All experimental protocols are provided.
Limitations: Tools for statistical analysis are not yet functional. Data are not downloadable
for offline analysis. Analytical reference libraries are not provided. Peak assignments are not
seamlessly integrated into the raw data viewer. No direct links between statistical results
and raw data vizualisation. Interface is not very intuitive.
Reference: (Ferry-Dumazet et al., 2011)
URL: http://www.cbib.u-bordeaux2.fr/MERYB/home/home.php

5.2.7 MetabolomeExpress
Description: An interactive, centralized metabolomics data repository for metabolomics
data from all organisms and all analytical platforms that provides a variety of cross-study
data-mining tools for analysis of metabolic phenotypes. Processed data may be uploaded in
a simple tab-delimited format. Alternatively, raw GC/MS data may be uploaded and
Online Metabolomics Databases and Pipelines 69

processing online using the integrated data-processing pipeline before being imported into
the data repository.
Species: Not formally species-constrained but current content is plant-centric. Data from
other systems is currently being gathered from the literature.
Reference data: MSRI libraries, GC/MS chromatograms, processed results, metadata in
systematic formats. Database currently includes >12000 publicly available metabolite
response statistics representing >100 metabolic phenotypes from 8 species under 22 different
experiments in 16 different peer-reviewed publications.
Noteworthy features: Members may independently upload their own MSRI libraries for
interactive dissemination and use within the GC/MS data-processing pipeline. Provides
tools for cross-study meta-analysis and database-driven phenotype recognition by pattern
matching.
Modes of access: browse and FTP
Strengths: All public data free for download. Provides a built-in GC/MS data processing
pipeline. Allows cross-study analysis. Processed metabolite response statistics are
transparently linked to underlying raw data in an interactive raw data viewer.
Limitations: No API. No search. Raw data processing pipeline needs to be extended to
support analytical platforms other than GC/MS. Does not provide as many multivariate
analysis and classification tools as other web-based metabolomics data-processing systems.
Reference: (Carroll et al., 2010)
URL: http://www.metabolome-express.org

6. Conclusion
The field of metabolomics informatics development is moving very rapidly. New data-
processing tools and new data repositories will continue to emerge. As they do, an
increasingly important area to make progress in will be in the standardization of universal
data exchange formats that allow free flow of data between compliant databases. Similarly
important will be the development of user-friendly metadata capture tools that make
systematic annotation of their datasets as painless as possible for biologists. These
developments will require the development of new ontologies and/or the extension of
existing ontologies that do not cover all of the terms required to describe metabolomics
experiments. The efficient sharing and mining of well-annotated and well-quality-controlled
metabolomics data across the internet will undoubtedly lead to many important discoveries
in the future.

7. References
Bais P, Moon SM, He K, Leitao R, Dreher K, Walk T, Sucaet Y, Barkan L, Wohlgemuth G,
Roth MR, Wurtele ES, Dixon P, Fiehn O, Lange BM, Shulaev V, Sumner LW, Welti
R, Nikolau BJ, Rhee SY, Dickerson JA (2010) PlantMetabolomics.org: a web portal
for plant metabolomics experiments. Plant physiology 152: 1807-1816
70 Metabolomics

Biswas A, Mynampati KC, Umashankar S, Reuben S, Parab G, Rao R, Kannan VS, Swarup S
(2010) MetDAT: a modular and workflow-based free online pipeline for mass
spectrometry data processing, analysis and interpretation. Bioinformatics 26: 2639-
2640
Carroll AJ, Badger MR, Harvey Millar A (2010) The MetabolomeExpress Project: enabling
web-based processing, analysis and transparent dissemination of GC/MS
metabolomics datasets. BMC Bioinformatics 11: 376
Caspi R, Altman T, Dale JM, Dreher K, Fulcher CA, Gilham F, Kaipa P, Karthikeyan AS,
Kothari A, Krummenacker M, Latendresse M, Mueller LA, Paley S, Popescu L,
Pujar A, Shearer AG, Zhang P, Karp PD (2010) The MetaCyc database of metabolic
pathways and enzymes and the BioCyc collection of pathway/genome databases.
Nucleic acids research 38: D473-479
Cochrane G, Karsch-Mizrachi I, Nakamura Y (2011) The International Nucleotide Sequence
Database Collaboration. Nucleic acids research 39: D15-D18
Croft D, O'Kelly G, Wu G, Haw R, Gillespie M, Matthews L, Caudy M, Garapati P, Gopinath
G, Jassal B, Jupe S, Kalatskaya I, Mahajan S, May B, Ndegwa N, Schmidt E,
Shamovsky V, Yung C, Birney E, Hermjakob H, D'Eustachio P, Stein L (2011)
Reactome: a database of reactions, pathways and biological processes. Nucleic
acids research 39: D691-697
Cui Q, Lewis IA, Hegeman AD, Anderson ME, Li J, Schulte CF, Westler WM, Eghbalnia HR,
Sussman MR, Markley JL (2008) Metabolite identification via the Madison
Metabolomics Consortium Database. Nature biotechnology 26: 162-164
Degtyarenko K, de Matos P, Ennis M, Hastings J, Zbinden M, McNaught A, Alcantara R,
Darsow M, Guedj M, Ashburner M (2008) ChEBI: a database and ontology for
chemical entities of biological interest. Nucleic acids research 36: D344-350
Ferry-Dumazet H, Gil L, Deborde C, Moing A, Bernillon S, Rolin D, Nikolski M, de Daruvar
A, Jacob D (2011) MeRy-B: a web knowledgebase for the storage, visualization,
analysis and annotation of plant NMR metabolomic profiles. BMC plant biology 11:
104
Hagiwara T, Saito S, Ujiie Y, Imai K, Kakuta M, Kadota K, Terada T, Sumikoshi K, Shimizu
K, Nishi T (2010) HPLC Retention time prediction for metabolome analysi.
Bioinformation 5: 255-258
Horai H, Arita M, Kanaya S, Nihei Y, Ikeda T, Suwa K, Ojima Y, Tanaka K, Tanaka S,
Aoshima K, Oda Y, Kakazu Y, Kusano M, Tohge T, Matsuda F, Sawada Y, Hirai
MY, Nakanishi H, Ikeda K, Akimoto N, Maoka T, Takahashi H, Ara T, Sakurai N,
Suzuki H, Shibata D, Neumann S, Iida T, Funatsu K, Matsuura F, Soga T, Taguchi
R, Saito K, Nishioka T (2010) MassBank: a public repository for sharing mass
spectral data for life sciences. Journal of mass spectrometry : JMS 45: 703-714
Iijima Y, Nakamura Y, Ogata Y, Tanaka Ki, Sakurai N, Suda K, Suzuki T, Suzuki H, Okazaki
K, Kitayama M, Kanaya S, Aoki K, Shibata D (2008) Metabolite annotations based
on the integration of mass spectral information. The Plant Journal 54: 949-962
Kaminuma E, Mashima J, Kodama Y, Gojobori T, Ogasawara O, Okubo K, Takagi T,
Nakamura Y (2010) DDBJ launches a new archive database with analytical tools for
next-generation sequence data. Nucleic acids research 38: D33-D38
Online Metabolomics Databases and Pipelines 71

Kopka J, Schauer N, Krueger S, Birkemeyer C, Usadel B, Bergmuller E, Dormann P,


Weckwerth W, Gibon Y, Stitt M, Willmitzer L, Fernie AR, Steinhauser D (2005)
GMD@CSB.DB: the Golm Metabolome Database. Bioinformatics 21: 1635-1638
Leinonen R, Sugawara H, Shumway M (2011) The Sequence Read Archive. Nucleic acids
research 39: D19-D21
Matsuda F, Hirai MY, Sasaki E, Akiyama K, Yonekura-Sakakibara K, Provart NJ, Sakurai T,
Shimada Y, Saito K (2010) AtMetExpress Development: A Phytochemical Atlas of
Arabidopsis Development. Plant Physiology 152: 566-578
Matsuda F, Yonekura-Sakakibara K, Niida R, Kuromori T, Shinozaki K, Saito K (2009)
MS/MS spectral tag-based annotation of non-targeted profile of plant secondary
metabolites. Plant J 57: 555-577
Moco S, Bino RJ, Vorst O, Verhoeven HA, de Groot J, van Beek TA, Vervoort J, de Vos CH
(2006) A liquid chromatography-mass spectrometry-based metabolome database
for tomato. Plant Physiology 141: 1205-1218
Neuweger H, Albaum SP, Dondrup M, Persicke M, Watt T, Niehaus K, Stoye J, Goesmann A
(2008) MeltDB: a software platform for the analysis and integration of
metabolomics experiment data. Bioinformatics 24: 2726-2732
Ogata H, Goto S, Sato K, Fujibuchi W, Bono H, Kanehisa M (1999) KEGG: Kyoto
Encyclopedia of Genes and Genomes. Nucleic acids research 27: 29-34
Sakurai N, Ara T, Ogata Y, Sano R, Ohno T, Sugiyama K, Hiruta A, Yamazaki K, Yano K,
Aoki K, Aharoni A, Hamada K, Yokoyama K, Kawamura S, Otsuka H, Tokimatsu
T, Kanehisa M, Suzuki H, Saito K, Shibata D (2010) KaPPA-View4: a metabolic
pathway database for representation and analysis of correlation networks of gene
co-expression and metabolite co-accumulation and omics data. Nucleic acids
research
Sansone SA, Fan T, Goodacre R, Griffin JL, Hardy NW, Kaddurah-Daouk R, Kristal BS,
Lindon J, Mendes P, Morrison N, Nikolau B, Robertson D, Sumner LW, Taylor C,
van der Werf M, van Ommen B, Fiehn O (2007) The metabolomics standards
initiative. Nature biotechnology 25: 846-848
Schauer N, Steinhauser D, Strelkov S, Schomburg D, Allison G, Moritz T, Lundgren K,
Roessner-Tunali U, Forbes MG, Willmitzer L, Fernie AR, Kopka J (2005) GC-MS
libraries for the rapid identification of metabolites in complex biological samples.
FEBS Lett 579: 1332-1337
Scholz M, Fiehn O (2007) SetupX--a public study design database for metabolomic projects.
Pacific Symposium on Biocomputing. Pacific Symposium on Biocomputing: 169-
180
Shinbo Y, Nakamura Y, Altaf-Ul-Amin M, Asahi H, Kurokawa K, Arita M, Saito K, Ohta D,
Shibata D, Kanaya S (2006) KNApSAcK: A Comprehensive Species-Metabolite
Relationship Database. In K Saito, RA Dixon, L Willmitzer, eds, Plant
Metabolomics, Vol 57. Springer Berlin Heidelberg, pp 165-181
Skogerson K, Wohlgemuth G, Barupal DK, Fiehn O (2011) The volatile compound BinBase
mass spectral database. BMC Bioinformatics 12: 321
Smith CA, O'Maille G, Want EJ, Qin C, Trauger SA, Brandon TR, Custodio DE, Abagyan R,
Siuzdak G (2005) METLIN: a metabolite mass spectral database. Therapeutic drug
monitoring 27: 747-751
72 Metabolomics

Smith CA, Want EJ, O'Maille G, Abagyan R, Siuzdak G (2006) XCMS: processing mass
spectrometry data for metabolite profiling using nonlinear peak alignment,
matching, and identification. Analytical chemistry 78: 779-787
Tarasova IA, Guryca V, Pridatchenko ML, Gorshkov AV, Kieffer-Jaquinod S, Evreinov VV,
Masselon CD, Gorshkov MV (2009) Standardization of retention time data for AMT
tag proteomics database generation. Journal of Chromatography B 877: 433-440
Ulrich EL, Akutsu H, Doreleijers JF, Harano Y, Ioannidis YE, Lin J, Livny M, Mading S,
Maziuk D, Miller Z, Nakatani E, Schulte CF, Tolmie DE, Kent Wenger R, Yao H,
Markley JL (2008) BioMagResBank. Nucleic acids research 36: D402-408
Wishart DS, Tzur D, Knox C, Eisner R, Guo AC, Young N, Cheng D, Jewell K, Arndt D,
Sawhney S, Fung C, Nikolai L, Lewis M, Coutouly MA, Forsythe I, Tang P,
Shrivastava S, Jeroncic K, Stothard P, Amegbey G, Block D, Hau DD, Wagner J,
Miniaci J, Clements M, Gebremedhin M, Guo N, Zhang Y, Duggan GE, Macinnis
GD, Weljie AM, Dowlatabadi R, Bamforth F, Clive D, Greiner R, Li L, Marrie T,
Sykes BD, Vogel HJ, Querengesser L (2007) HMDB: the Human Metabolome
Database. Nucleic acids research 35: D521-526
Wohlgemuth G, Haldiya PK, Willighagen E, Kind T, Fiehn O (2010) The Chemical
Translation Service--a web-based tool to improve standardization of metabolomic
reports. Bioinformatics 26: 2647-2648
Xia J, Psychogios N, Young N, Wishart DS (2009) MetaboAnalyst: a web server for
metabolomic data analysis and interpretation. Nucleic acids research 37: W652-660
0
4

Generic Software Frameworks for


GC-MS Based Metabolomics
Nils Hoffmann and Jens Stoye
Genome Informatics, Faculty of Technology, Bielefeld University
Germany

1. Introduction
Metabolomics has seen a rapid development of new technologies, methodologies, and
data analysis procedures during the past decade. The development of fast gas- and
liquid-chromatography devices coupled to sensitive mass-spectrometers, supplemented by
the unprecedented precision of nuclear magnetic resonance for structure elucidation of
small molecules, together with the public availability of database resources associated to
metabolites and metabolic pathways, has enabled researchers to approach the metabolome
of organisms in a high-throughput fashion. Other "omics" technologies have a longer history
in high-throughput, such as next generation sequencing for genomics, RNA microarrays for
transcriptomics, and mass spectrometry methods for proteomics. All of these together give
researchers a unique opportunity to study and combine multi-omics aspects, forming the
discipline of "Systems Biology" in order to study organisms at multiple scales simultaneously.
Like all other "omics" technologies, metabolomics data acquisition is becoming more reliable
and less costly, while at the same time throughput is increased. Modern time-of-flight (TOF)
mass spectrometers are capable of acquiring full scan mass spectra at a rate of 500Hz from 50
to 750 m/z and with a mass accuracy <5 ppm with external calibration (Neumann & Böcker,
2010). At the opposite extreme of machinery, Fourier-transform ion-cyclotron-resonance
(FTICR) mass spectrometers coupled to liquid chromatography for sample separation reach
an unprecedented mass accuracy of <1 ppm m/z and very high mass resolution (Miura
et al., 2010). These features are key requirements for successful and unique identification of
metabolites. Coupled to chromatographic separation devices, these machines create datasets
ranging in size from a few hundred megabytes to several gigabytes per run. While this is not
a severe limitation for small scale experiments, it may pose a significant burden on projects
that aim at studying the metabolome or specific metabolites of a large number of specimens
and replicates, for example in medical research studies or in routine diagnostics applications
tailored to the metabolome of a specific species (Wishart et al., 2009).
Thus, there is a need for sophisticated methods that can treat these datasets efficiently in terms
of computational resources and which are able to extract, process, and compare the relevant
information from these datasets. Many such methods have been published, however there is
a high degree of fragmentation concerning the availability and accessibility of these methods,
which makes it hard to integrate them into a lab’s workflow.
The aim of this work is to discuss the necessary and desirable features of a software framework
for metabolomics data preprocessing based on gas-chromatography (GC) and comprehensive
74
2 Metabolomics
Will-be-set-by-IN-TECH

two-dimensional gas-chromatography (GCxGC) coupled to single-dimension detectors


(flame/photo ionization, FID/PID) or multi-dimension detectors (mass spectrometry, MS).
We compare the features of publicly available Open Source frameworks that usually have
a steep learning curve for end-users and bioinformaticians alike, owing to their inherent
complexity. Many users will thus be appaled by the effort it takes to get used to a framework.
Thus, the main audience of this work are bioinformaticians and users willing to invest some
time in learning to use and/or program in these frameworks in order to set up a lab specific
analytical platform. For a review of LC-MS based metabolomics data preprocessing consider
(Castillo, Mattila, Miettinen, Orešič & Hyötyläinen, 2011).
Before we actually compare the capabilities of these different frameworks, we will first define
a typical workflow for automatic data processing of metabolomics experiments and will
discuss available methods within each of the workflow’s steps.
We will concentrate on frameworks available under an Open Source license, thus allowing
researchers to examine their actual implementation details. This distinguishes these
frameworks from applications that are only provided on explicit request, under limited terms
of use, or that are not published together with their source code (Lommen, 2009; Stein, 1999),
which is still often the case in metabolomics and may hamper comparability and reuse of
existing solutions. Additionally, all frameworks compared in this work are available for
all major operating systems such as Microsoft Windows, Linux, and Apple Mac OSx as
standalone applications or libraries.
Web-based methods are not compared within this work as they most often require a complex
infrastructure to be set up and maintained. However, we will give a short overview of recent
publications on this topic and provide short links to the parts of the metabolomics pipeline
that we discuss in the following section. A survey of web-based methods is provided by
Tohge & Fernie (2009). More recent web-based applications for metabolomics include the
retention time alignment methods Warp2D (Ahmad et al., 2011) and ChromA (Hoffmann &
Stoye, 2009), which are applicable to GC-MS or LC-MS data, and Chromaligner (Wang et al.,
2010), which aligns GC and LC data with single-dimension detectors like FID.
Tools for statistical analysis of multiple sample groups and with different phenotypes have
been reported by Kastenmüller et al. (2011). However, other tools aim to integrate a
more complete metabolomics workflow including preprocessing, peakfinding, alignment and
statistical analysis combined with pathway mapping information like MetaboAnalyst (Xia &
Wishart, 2011), MetabolomeExpress (Carroll et al., 2010), or MeltDB (Neuweger et al., 2008).
These larger web-based frameworks integrate other functionality for time-course analysis (Xia
et al., 2011), pathway mapping (Neuweger et al., 2009; Xia & Wishart, 2010a) and metabolite
set enrichment analysis (Kankainen et al., 2011; Xia & Wishart, 2010b).
In the Application section, we will exemplarily describe two pipelines for metabolomics
analyses based on our own Open Source framework Maltcms: ChromA, which is applicable
to GC-MS, and ChromA4D, which is applicable to data from comprehensive GCxGC-MS
experiments. We show how to set up, configure and execute each pipeline using instructional
datasets. These two workflows include the typical steps of raw-data preprocessing in
metabolomics, including peak-finding and integration, peak-matching among multiple
replicate groups and tentative identification using mass-spectral databases, as well as
visualizations of raw and processed data. We will describe the individual steps of the
753
Generic Software Frameworks for
Generic Software Frameworks for GC-MS Based Metabolomics
GC-MS Based Metabolomics

workflows of the two application pipelines to give the reader a thorough understanding of
the methods used by ChromA and ChromA4D.
Finally, we discuss the current state of the presented Open Source frameworks and give an
outlook into the future of software frameworks and data standards for metabolomics.

2. A typical workflow for a metabolomics experiment


Metabolomics can be defined as the study of the metabolic state of an organism or its response
to direct or indirect perturbation. In order to find differences between two or more states,
for example before treatment with a drug and after, and among one or multiple specimens,
the actual hypothesis for the experiment needs to be defined. Based on this hypothesis, a
design for the structure of the experiments and their subsequent analysis can be derived.
This involves, among many necessary biological or medical considerations, the choice of
sample extraction procedures and preparation methods, as well as the choice of the analytical
methods used for downstream sample analysis.
Preprocessing of the data from those experiments begins after the samples have been acquired
using the chosen analytical method, such as GC-MS or LC-MS. Owing to the increasing
amount of data produced by high-throughput metabolomics experiments, with large sample
numbers and high-accuracy/high-speed analytical devices, it is a key requirement that the
resulting data is processed with very high level of automation. It is then that the following
typical workflow is applied in some variation, as illustrated in Figure 1.

Sample Collection Interpretation

Sample Preparation Statistical Evaluation

Data Acquisition and Conversion Alignment

Preprocessing Peak Detection

Fig. 1. A typical workflow for a metabolomics experiment. Steps shown in orange (solid
border) are usually handled within the bioinformatics domain, while the steps shown in
green (dashed border) often involve co-work with scientists from other disciplines.

2.1 Data acquisition and conversion


The most common formats exported from GC-MS and LC-MS machines today are NetCDF
(Rew & Davis, 1990), based on the specifications in the ASTM/AIA standard ANDI-MS
(Matthews, 2000), mzXML (Oliver et al., 2004), mzData (Orchard et al., 2005), and more
76
4 Metabolomics
Will-be-set-by-IN-TECH

recently as the successor to the latter two, mzML (Deutsch, 2008; Martens et al., 2010). All of
these formats include well-defined data structures for meta-information necessary to interpret
data in the right context, such as detector type, chromatographic protocol, detector potential
and other details about the separation and acquisition of the data. Furthermore, they explicitly
model chromatograms and mass spectra, with varying degrees of detail.
NetCDF is the oldest and probably most widely used format today. It is routinely
exported even by older machinery, which offers backwards compatibility to those. It is
a general-purpose binary format, with a header that describes the structure of the data
contained in the file, grouped into variables and indexed by dimensions. In recent years,
efforts were made to establish open formats for data exchange based on a defined grammar
in extensible markup language (XML) with extendable controlled vocabularies, to allow
new technologies to be easily incorporated into the file format without breaking backwards
compatibility. Additionally, XML formats are human readable which narrows the technology
gap. mzXML was the first approach to establish such a format. It has been superseded
by mzData and, more recently, mzML was designed as a super-set of both, incorporating
extensibility through the use of an indexed controlled vocabulary. This allows mzML to be
adapted to technologies like GCxGC-MS without having to change its definition, although
its origins are in the proteomics domain. One drawback of XML-based formats is often
claimed to be their considerably larger space requirements when compared to the supposedly
more compact binary data representations. Recent advances in mzML approach this issue by
compressing spectral data using gzip compression.
The data is continuously stored in a vendor-dependent native format during sample
processing on a GC-MS machine. Along with the mass spectral information, like ion mass (or
equivalents) and abundance, the acquisition time of each mass spectrum is recorded. Usually,
the vendor software includes methods for data conversion into one of the aforementioned
formats. However, especially when a high degreee of automation is desired, it may be
beneficial to directly access the data in their native format. This avoids the need to run
the vendor’s proprietary software manually for every data conversion task. Both the
ProteoWizard framework (Kessner et al., 2008) and the Trans Proteomic Pipeline (Deutsch
et al., 2010) include multiple vendor-specific libraries for that use case.

2.2 Preprocessing
Raw mass specrometry data is usually represented in sparse formats, only recording those
masses whose intensities exceed a user-defined threshold. This thresholding is usually
applied within the vendor’s proprietary software and may lead to artificial gaps within the
data. Thus, the first step in preprocessing involves the binning of mass spectra over time
into bins of defined size in the m/z dimension, followed by interpolation of missing values.
After binning, the data is stored as a rectangular array of values, with the first dimension
representing time, the second dimension representing the approximate bin mass values, and
the third dimension representing the intensity corresponding to each measured ion. This
process is also often described as resampling (Lange et al., 2007).
Depending on various instrumental parameters, the raw exported data may require additional
processing. The most commonly reported methods for smoothing are the Savitzky-Golay
filter (Savitzky & Golay, 1964), LOESS regression (Smith et al., 2006) and variants of local
averaging, for example by a windowed moving average filter. These methods can also be
Generic
GC-MS BasedSoftware
Metabolomics Frameworks for GC-MS Based Metabolomics 775
Generic Software Frameworks for

applied to interpolate values where gaps are present in the original data. The top-hat filter
(Bertsch et al., 2008; Lange et al., 2007) is used to remove a varying baseline from the signal.
More refined methods use signal decomposition and reconstruction methods, such as Fourier
transform and continuous wavelet transform (CWT) (Du et al., 2006; Fredriksson et al., 2009;
Tautenhahn et al., 2008) in order to remove noise and baseline contributions from the signal
and simultaneously find peaks.

2.3 Peak detection


Often the process of peak detection is decoupled from the actual preprocessing of the data.
XCMS (Smith et al., 2006), for example, uses a Gaussian second derivative peak model with
a fixed kernel width and signal-to-noise threshold to find peaks along the chromatographic
domain of each ion bin. Other methods extend this approach to use a multi-scale continuous
wavelet transform using such a kernel over various widths, tracking the response of the
transformed signal in order to locate peak apex positions in scale-space before estimating
the true peak widths based on the kernel scale with maximum response (Fredriksson et al.,
2009; Tautenhahn et al., 2008). However, these methods usually allow only a small number of
co-eluting peaks in different mass-bins, since they were initially designed to work with LC-MS
data mainly, where only one parent ion and a limited number of accompanying adduct ions
are expected. In GC-MS, electron-ionization creates rich fragmentation mass spectra, which
pose additional challenges to deconvolution of co-eluting ions and subsequent association
to peak groups. Even though its source code is not publicly available, the method used by
AMDIS (Stein, 1999) has seen wide practical application and is well accepted as a reference by
the metabolomics and analytical chemistry communities.

2.4 Alignment
The alignment problem in metabolomics and proteomics stems from the analytical
methods used. These produce sampled sensor readings acquired over time in fixed or
programmed intervals, usually called chromatograms. The sensor readings can be one- or
multidimensional. In the first case, detectors like ultra violet and visible light absorbance
detectors (UV/VIS) or flame ionization detectors (FID) measure the signal response as
one-dimensional features, e.g. as the absorbance spectrum or electrical potential, respectively.
Multi-dimensional detectors like mass spectrometers record a large number of features
simultaneously, e.g. mass and ion count. The task is then to find corresponding and
non-corresponding features between different sample acquisitions. This correspondence
problem is a term used by Åberg et al. (2009) which describes the actual purpose of alignment,
namely to find true correspondences between related analytical signals over a number of
sample acquisitions. For GC-MS- and LC-MS-based data, a number of different methods have
been developed, some of which are described in more detail by Castillo, Gopalacharyulu,
Yetukuri & Orešič (2011) and Åberg et al. (2009). Here, we will concentrate on those methods
that have been reported to be applicable to GC-MS. In principle, alignment algorithms can
be classified into two main categories: peak- and signal-based methods. Methods of the first
type start with a defined set of peaks, which are present in most or all samples that are to be
aligned before determining the best correspondences of the peaks between samples in order to
then derive a time correction function. Krebs et al. (2006) locate landmark peaks in the TIC and
then select pairs of those peaks with a high correlation between their mass spectra in order to
fit an interpolating spline between a reference chromatogram and the to-be-aligned one. The
78
6 Metabolomics
Will-be-set-by-IN-TECH

method of Robinson et al. (2007) is inspired by multiple sequence alignment algorithms and
uses dynamic programming to progressively align peak lists without requiring an explicit
reference chromatogram. Other methods, like that of Chae et al. (2008) perform piecewise,
block-oriented matching of peaks, either on the TIC, on selected masses, or on the complete
mass spectra. Time correction is applied after the peak assignments between the reference
chromatogram and the others have been calculated. Signal-based methods include recent
variants of correlation optimized warping (Smilde & Horvatovich, 2008), parametric time
warping (Christin et al., 2010) and dynamic time warping (Christin et al., 2010; Clifford et al.,
2009; Hoffmann & Stoye, 2009; Prince & Marcotte, 2006) and usually consider the complete
chromatogram for comparison. However, attempts are made to reduce the computational
burden associated with a complete pairwise comparison of mass spectra by partitioning the
chromatograms into similar regions (Hoffmann & Stoye, 2009), or by selecting a representative
subset of mass traces (Christin et al., 2010). Another distinction in alignment algorithms is the
requirement of an explicit reference for alignment. Some methods apply clustering techniques
to select one chromatogram that is most similar to all others (Hoffmann & Stoye, 2009; Smilde
& Horvatovich, 2008), while other methods choose such a reference based on the number of
features contained in a chromatogram (Lange et al., 2007) or by manual user choice (Chae
et al., 2008; Clifford et al., 2009). For high-throughput applications, alignments should be fast
to calculate and reference selection should be automatic. Thus, a sampling method for time
correction has recently been reported by Pluskal et al. (2010) for LC-MS. A comparison of these
methods is given in the same publication.

2.5 Statistical evaluation


After peaks have been located and integrated for all samples, and their correspondence has
been established, peak report tables can be generated, containing peak information for each
sample and peak, with associated corrected retention times and peak areas. Additionally,
peaks may have been putatively identified by searching against a database, such as the GMD
(Hummel et al., 2007) or the NIST mass-spectral database (Babushok et al., 2007).
These peak tables can then be analyzed with further methods, in order to detect e.g. systematic
differences between different sample groups. Prior to such an analysis, the peak areas need
to be normalized. This is usually done by using a spiked-in compound which is not expected
to occur naturally as a reference. The normalization compound is supposed to have the same
concentration in all samples. The compound’s peak area can then be used to normalize all
peak areas of a sample with respect to it (Doebbe et al., 2010).
Different experimental designs allow to analyze correlations of metabolite levels for the same
subjects under different conditions (paired), or within and between groups of subjects. For
simple paired settings, multiple t-tests with corrections for multiple testing can be applied
(Berk et al., 2011), while for comparisons between groups of subjects, Fisher’s F-Statistic
(Pierce et al., 2006) and various analysis of variance (ANOVA), principal component analysis
(PCA) and partial least squares (PLS) methods are applied (Kastenmüller et al., 2011; Wiklund
et al., 2008; Xia et al., 2011).

2.6 Evaluation of hypothesis


Finally, after peak areas have been normalized and differences have been found between
sample groups, the actual results need to be put into context and be interpreted in their
Generic
GC-MS BasedSoftware
Metabolomics Frameworks for GC-MS Based Metabolomics 797
Generic Software Frameworks for

biological context. This task is usually not handled by the frameworks described in this
chapter. Many web-based analysis tools allow to put the data into a larger context,
by providing name- or id-based mapping of the experimentally determined metabolite
concentrations onto biochemical pathways like MetaboAnalyst (Xia & Wishart, 2011),
MetabolomeExpress (Carroll et al., 2010), or MeltDB (Neuweger et al., 2008). The latter allows
association of the metabolomics data with other results for the same subjects under study or
with results from other "omics" experiments on the same target subjects, but this is beyond
the scope of the frameworks presented herein.

3. Frameworks for GC-MS analysis


A number of Open Source frameworks have been developed for LC-MS based proteomics
frameworks like OpenMS (Bertsch et al., 2008), ProteoWizard (Kessner et al., 2008), and
most notably the TransProteomicPipeline (Deutsch et al., 2010). Even though many of
the steps required for proteomics apply similarily to metabolomics applications, there are
still some essential differences due to the different analytical setups and technologies (e.g.
matrix assisted laser desorption ionization mass spectrometry, MALDI-MS) used in the two
fields. XCMS (Smith et al., 2006) was among the first frameworks to offer support for data
preprocessing in LC-MS based metabolomics. Later, MZmine2 (Pluskal et al., 2010) offered an
alternative with a user-friendly interface and easy extendability. Lately, Scheltema et al. (2011)
published their PeakML format and mzMatch framework also for LC-MS applications. As
of now, there seem to be only a few frameworks available for GC-MS based metabolomics
that offer similar methods, namely PyMS (Callaghan et al., 2010; Isaac et al., 2009) and
Maltcms/ChromA (Hoffmann & Stoye, 2009; Maltcms, 2011) . These will be presented in more
detail in this section. A compact overview of the Open Source frameworks discussed herein
is given in Table 1. A detailed feature comparison can be found in Table 2.

3.1 XCMS
XCMS (Smith et al., 2006) is a very mature framework and has seen constant development
during the last five years. It is mainly designed for LC-MS applications, however its binning,
peak finding and alignment are also applicable to GC-MS data. XCMS is implemented in
the GNU R programming language, the de-facto standard for Open Source statistics. Since
GNU R is an interpreted scripting language, it is easy to write custom scripts that realize
additional functionality of the typical GC-MS workflow described above. XCMS is part of
the Bioconductor package collection, which offers many computational methods for various
"omics" technologies. Further statistical methods are available from GNU R.
XCMS supports input in NetCDF, mzXML, mzData and, more recently, mzML format. This
allows XCMS to be used with virtually any chromatography-mass spectrometry data, since
vendor software supports conversion to at least one of those formats. XCMS uses the xcmsRaw
object as its primary tabular data structure for each binned data file. The xcmsSet object is then
used to represent peaks and peak groups and is used by its peak alignment and diffreport
features.
The peak finding methods in XCMS are quite different from each other. For data with normal
or low mass resolution and accuracy, the matched filter peak finder (Smith et al., 2006)
is usually sensitive enough. It uses a Gaussian peak template function with user defined
width and signal-to-noise critera to locate peaks on individual binned extracted ion current
80
8 Metabolomics
Will-be-set-by-IN-TECH

(EIC) traces over the complete time range of the binned chromatogram. The other method,
CentWave (Tautenhahn et al., 2008) is based on a continuous wavelet transform on areas of
interest within the raw data matrix. Both peak finding methods report peak boundaries and
integrated areas for raw data and for the data reconstructed from the peak finder’s signal
response values.
Initially designed for LC-MS, XCMS does not have a method to group co-eluting peaks into
peak groups, as is a requirement in GC-MS methods using electron ionization. However,
CAMERA (Tautenhahn et al., 2007) shows how XCMS can be used as a basis in order to create
a derived application, in this case for ion annotation between samples.
Peak alignment in XCMS is performed using local LOESS regression between peak groups
with very similar m/z and retention time behaviour and good support within each sample
group. This allows a simultaneous alignment and retention time correction of all peaks. The
other available method is based on the Obi-Warp dynamic time warping (Prince & Marcotte,
2006) algorithm and is capable of correcting large non-linear retention time distortions. It uses
the peak set with the highest number of features as alignment reference, which is comparable
to the approach used by Lange et al. (2007). However, it is much more computationally
demanding then the LOESS-based alignment.
XCMS’s diffreport generates a summary report of significant analyte differences between two
sample sets. It uses Welch’s two-sample t-statistic to calculate p-values for each analyte group.
ANOVA may be used for more than two sample sets.
A number of different visualizations are also available, both for raw and processed data. These
include TIC plots, EIC plots, analyte group plots for grouped features, and chromatogram (rt,
m/z, intensity) surface plots.
XCMS can use GNU R’s Rmpi infrastructure to execute arbitary function calls, such as profile
generation and peak finding, in parallel on a local cluster of computers.

3.2 PyMS
PyMS (Callaghan et al., 2010; Isaac et al., 2009) is a programming framework for GC-MS
metabolomics based on the Python programming language. It can therefore use a large
number of scientific libraries which are accessible via the SciPy and NumPy packages (SciPy,
2011). Since Python is a scripting language, it allows to do rapid prototyping, comparable to
GNU R. However, Python’s syntax may be more familiar for programmers with a background
in object-oriented programming languages.
The downloadable version of PyMS currently only supports NetCDF among the more recent
open data exchange formats. Nonetheless, it is the only framework in this comparison with
support for the JCAMP GC-MS file format.
PyMS provides dedicated data structures for chromatograms, allowing efficient access to
EICs, mass spectra, and peak data.
In order to find peaks, PyMS also builds a rectangular profile matrix with the dimensions
time, m/z and intensity. Through the use of slightly shifted binning boundaries, they
reduce the chance of false assignments of ion signals to neighboring bins, when binning is
performed with unit precision (bin width of 1 m/z). PyMS offers the moving average and
the Savitzky-Golay (Savitzky & Golay, 1964) filters for signal smoothing of EICs within the
Generic
GC-MS BasedSoftware
Metabolomics Frameworks for GC-MS Based Metabolomics 819
Generic Software Frameworks for

profile matrix. Baseline correction can be performed by the top-hat filter (Lange et al., 2007).
The actual peak finding is based on the method described by Biller & Biemann (1974) and
involves the matching of local peak maxima co-eluting within a defined window. Peaks are
integrated for all co-eluting masses, starting from a peak apex to both sides and ending if the
increase in area falls below a given threshold.
Peak alignment in PyMS is realized by the method introduced by Robinson et al. (2007). It
is related to progressive multiple sequence alignment methods and is based on a generic
dynamic programming algorithm for peak lists. It proceeds by first aligning peak lists within
sample groups, before aligning the aligned peak lists of different groups, until all groups have
been aligned.
Visualizations of chromatogram TICs, EICs, peaks and mass spectra are available and are
displayed to the user in an interactive plot panel.
For high-throughput applications, PyMS can be used together with MPI to parallelize tasks
within a local cluster of computers.

3.3 Maltcms
The framework Maltcms allows to set up and configure individual processing components
for various types of computational analyses of metabolomics data. The framework is
implemented in JAVA and is modular using the service provider pattern for maximal
decoupling of interface and implementation, so that it can be extended in functionality at
runtime.
Maltcms can read data from files in NetCDF, mzXML, mzData or mzML format. It uses a
pipeline paradigm to model the typical preprocessing workflow in metabolomics, where each
processing step can define dependencies on previous steps. This allows automatic pipeline
validation and ensures that a user can not define an invalid pipeline. The workflow itself is
serialized to XML format, keeping track of all resources created during pipeline execution.
Using a custom post-processor, users can define which results of the pipeline should be
archived.
Maltcms uses a generalization of the ANDI-MS data schema internally and a data provider
interface with corresponding implementations to perform the mapping from any proprietary
data format to an internal data object model. This allows efficient access to individual mass
spectra and other data available in the raw-data files. Additionally, developers need no special
knowledge of any supported file format, since all data can be accessed generically. Results
from previous processing steps are referenced in the data model to allow both shadowing of
data, e.g. creating a processing result variable with the same name as an already existing
variable, and aggregation of processing results. Thus, all previous processing results are
transparently accessible for downstream elements of a processing pipeline, unless they have
been shadowed.
Primary storage of processing results is performed on a per-chromatogram basis in the binary
NetCDF file format. Since metabolomics experiments create large amounts of data, a focus is
put on efficient data structures, data access, and scalability of the framework.
82
10 Metabolomics
Will-be-set-by-IN-TECH

Embedding Maltcms in existing workflows or interfacing with other software is also possible,
as alignments, peak-lists and other feature data can be exported as comma separated value
files or in specific xml-based formats, which are well-defined by custom schemas.
To exploit the potential of modern multi-core CPUs and distributed computing networks,
Maltcms supports multi-threaded execution on a local machine or within a grid of connected
computers using an OpenGrid infrastructure (e.g. Oracle Grid Engine or Globus Toolkit
(Foster, 2005)) or a manually connected network of machines via remote method invocation
(RMI).
The framework is accompanied by many libraries for different purposes, such as the
JFreeChart library for 2D-plotting or, for BLAS compatible linear algebra, math and statistics
implementations, the Colt and commons-math libraries. Building upon the base library Cross,
which defines the commonly available interfaces and default implementations, Maltcms
provides the domain dependent data structures and specializations for processing of
chromatographic data.

Name Version Analytical method Software license Programming language


XCMS 1.26.1a LC-MS/GC-MS GNU GPL v2 GNU R 2.13/C++
PyMS r371 GC-MS GNU GPL v2 Python 2.5
Maltcms/ChromA 1.1 GC-MS GNU L-GPL v3 JAVA 6

Table 1. Overview of available Open Source software frameworks for GC-MS based
metabolomics. a: Part of Bioconductor 2.8

Feature (GC-MS pipeline) XCMS PyMS ChromA


Data formats A, B, C, D A, E A, B, C, D
Signal preprocessing MM SG, TH MA, MM, TH
Peak detection MF, CWT BB MAX
Multiple peak alignment LOESS, DTW PROGDP DTW, CLIQUE
Visualization TIC, EIC, SURF TIC, EIC TIC, EIC, SURF
DB search no (LC-MS only) no MSP
Normalization no no RP, EV
Statistical evaluation TT no FT

Table 2. Feature comparison of Open Source software frameworks for preprocessing of


GC-MS based metabolomics data. Keys to abbreviations: Data formats A: NetCDF, B:
mzXML, C: mzData, D: mzML, E: JCAMP GC-MS. Signal preprocessing MM: moving
median, SG: Savitzky-Golay filter, TH: top-hat filter, MA: moving average. Peak detection
MF: matched Gaussian filter, CWT: continuous wavelet transform, BB: Biller-Biemann, MAX:
TIC local maxima. Multiple peak alignment LOESS: LOESS regression, DTW: dynamic time
warping, PROGDP: progressive using dynamic programming, CLIQUE: progressive
clique-based. Visualization (of unaligned and aligned data) TIC: plots of total ion
chromatogram/peaks, EIC: plots of extracted ion chromatograms/peaks, SURF: surface
plots of profile matrix (rt x m/z x I). DB search MSP: msp-format, compatible with AMDIS
and GMD format. Normalization RP: reference peak area, EV: external value, e.g. dry
weight. Statistical evaluation TT: groupwise t-test, multiple testing correction, FT: F-test,
between group vs. within group variance
Generic
GC-MS BasedSoftware
Metabolomics Frameworks for GC-MS Based Metabolomics 83
Generic Software Frameworks for
11

3.3.1 ChromA
ChromA is a configuration of Maltcms that includes preprocessing, in the form of mass
binning, time-scale alignment and annotation of signal peaks found within the data, as
well as visualizations of unaligned and aligned data from GC-MS and LC-MS experiments.
The user may supply mandatory alignment anchors as CSV files to the pipeline and a
database location for tentative metabolite identification. Further downstream processing can
be performed either on the retention time-corrected chromatograms in NetCDF format, or on
the corresponding peak tables in either CSV format or XML format.
Peaks can either be imported from other tools, by providing them in CSV format to ChromA,
giving at least the scan index of each peak in a file per row. Alternatively, ChromA has a fast
peak finder that locates peaks based on derivatives of the smoothed and baseline-corrected
TIC, using a moving average filter followed by top-hat filter baseline-substraction, with a
predefined minimum peak-width. Peak alignment is based on a star-wise or tree-based
application of an enhanced variant of pairwise dynamic time warping (DTW) (Hoffmann &
Stoye, 2009). To reduce both runtime and space requirements, conserved signals throughout
the data are identified, constraining the search space of DTW to a precomputed closed
polygon. The alignment anchors can be augmented or overwritten by user-defined anchors,
such as previously identified compounds, characteristic mass or MS/MS identifications.
Then, the candidates are paired by means of a bidirectional best-hits (BBH) criterion, which
can compare different aspects of the candidates for similarity. Paired anchors are extended to
k-cliques with configurable k, which help to determine the conservation or absence of signals
across measurements, especially with respect to replicate groups. Tentative identification
of peaks against a database using their mass spectra is possible using the MetaboliteDB
module. This module provides access to mass-spectral databases in msp-compatible format,
for example the Golm Metabolite Database or the NIST EI-MS database.
ChromA visualizes alignment results including paired anchors in birds-eye view or as a
simultaneous overlay plot of the TIC. Additionally, absolute and relative differential charts
are provided, which allow easy spotting of quantitative differences.
Peak tables are exported in CSV format, including peak apex positions, area under curve, peak
intensity and possibly tentative database identifications. Additionally, information about the
matched and aligned peak groups is saved in CSV format.

4. Frameworks for GCxGC-MS analysis


The automatic and routine analysis of comprehensive GCxGC-MS data is yet to be established.
GCxGC-MS couples a second chromatographic column to the first one, thereby achieving a
much higher peak capacity and thus a better separation of closely co-eluting analytes (Castillo,
Mattila, Miettinen, Orešič & Hyötyläinen, 2011). Usually, for a one-hour run, the raw data
file size exceeds a few Gigabytes. Quite a number of algorithms have been published on
alignment of peaks in such four-dimensional (first column retention time, second column
retention time, mass, and intensity values) data (Kim et al., 2011; Oh et al., 2008; Pierce et al.,
2005; Vial et al., 2009; Zhang, 2010), however only a few methods are available for a more
complete typical preprocessing workflow. A compact overview of the available frameworks,
their licenses and programming languages is given in Table 3. Table 4 gives a more detailed
feature matrix of these frameworks. The remainder of this section gives a concise overview
84
12 Metabolomics
Will-be-set-by-IN-TECH

of the frameworks Guineu (Castillo, Mattila, Miettinen, Orešič & Hyötyläinen, 2011) and
ChromA4D (Maltcms, 2011).

Name Version Supported methods Software license Programming language


Guineu 0.8.2 GCxGC-MS (LC-MS) GNU GPL v2 JAVA 6
Maltcms/ChromA4D 1.1 GCxGC-MS GNU L-GPL v3 JAVA 6

Table 3. Feature comparison of Open Source software frameworks for GCxGC-MS based
metabolomics

4.1 Guineu
Guineu is a recently published graphical user interface and application for the comparative
analysis of GCxGC-MS data (Castillo, Mattila, Miettinen, Orešič & Hyötyläinen, 2011). It
currently reads LECO ChromaTOF software’s peak list output after smoothing, baseline
correction, peak finding, deconvolution, database search and retention index (RI) calculation
have been performed within ChromaTOF.
The peak lists are aligned pairwise using the score alignment algorithm, which requires
user-defined retention time windows for both separation dimensions. Additionally, the
one-dimensional retention index (RI) of each peak is used within the score calculation. Finally,

Feature (GCxGC-MS pipeline) Guineu ChromA4D


Data formats G A,H
Signal preprocessing no MA, MM, TH, CV
Peak detection no MAX-SRG
Multiple peak alignment SCORE CLIQUE
Visualization STATS STATS, TIC, EIC, TIC2D
DB search GMD, PUBCHEM, KEGG MSP (GMD)
Normalization RP RP, EV
Statistical evaluation CV, FLT, TT, PCA, CDA, SP, ANOVA FT

Table 4. Feature comparison of Open Source software frameworks for preprocessing of


GCxGC-MS based metabolomics data. Key to abbreviations: Data formats A: NetCDF, G:
ChromaTOF peak lists, H: CSV peak lists. Signal preprocessing MA: moving average, MM:
moving median, TH: top-hat filter, CV: coefficient of variation threshold. Peak detection
MAX-SRG: TIC local maxima, seeded region growing based on ms similarity. Multiple peak
alignment SCORE: parallel iterative score-based, CLIQUE: progressive
clique-based.Visualization (of unaligned and aligned data) TIC: plots of total ion
chromatogram/peaks, EIC: plots of extracted ion chromatograms/peaks, SURF: surface
plots of profile matrix (rt x m/z x I), STATS: visualization of statistical values. DB search
GMD: Golm metabolite database webservice, PUBCHEM: pubchem database webservice,
KEGG: kegg metabolite database, MSP: msp-format, compatible with AMDIS and GMD
format. Normalization RP: reference peak area, EV: external value, e.g. dry weight.
Statistical evaluation CV: coefficient of variation, FLT: fold-test, TT: groupwise t-test, PCA:
principal components analysis, CDA: curvilinear distance analysis, SP: Sammon’s projection,
ANOVA: analysis of variance, FT: F-test, between group vs. within group variance.
Generic
GC-MS BasedSoftware
Metabolomics Frameworks for GC-MS Based Metabolomics 85
Generic Software Frameworks for
13

a threshold for mass spectral similarity is needed in order to create putative peak groups.
Additional peak lists are added incrementally to an already aligned path, based on the
individual peaks’ score against those peaks that are already contained within the path.
Guineu provides different filters to remove peaks by name, group occurrence count, or other
features from the ChromaTOF peak table. In order to identify compound classes, the Golm
metabolite database (GMD) substructure search is used. Peak areas can be extracted from
ChromaTOF using the TIC, or using extracted, informative or unique masses. Peak area
normalization is available relative to multiple user-defined standard compounds.
After peak list processing, Guineu produces an output table containing information for
all aligned peaks, containing information on the original analyte annotation as given by
ChromaTOF, peak areas, average retention times in both dimensions together with the average
RI and further chemical information on the functional group and substructure prediction as
given by the GMD. It is also possible to link the peak data to KEGG and Pubchem via the CAS
annotation, if it is available for the reported analyte.
For statistical analysis of the peak data, Guineu provides fold change- and t-tests, principal
component analysis (PCA), analysis of variance (ANOVA) and other methods.
Guineu’s statistical analysis methods provide different plots of the data sets, e.g. for showing
the principal components of variation within the data sets after analysis with PCA.

4.2 ChromA4D
For the comparison of comprehensive two-dimensional gas chromatography-mass
spectrometry (GCxGC-MS) data, ChromA4D accepts NetCDF files as input. Additionally,
the user needs to provide the total runtime on the second orthogonal column (modulation
time) to calculate the second retention dimension information from the raw data files. For
tentative metabolite identification, the location of a database can be given by the user.
ChromA4D reports the located peaks, their respective integrated TIC areas, their best
matching corresponding peaks in other chromatograms, as well as a tentative identification
for each peak. Furthermore, all peaks are exported together with their mass spectra to MSP
format, which allows for downstream processing and re-analysis with AMDIS and other
tools. The exported MSP files may be used to define a custom database of reference spectra
for subsequent analyses.
Peak areas are found by a modified seeded region growing algorithm. All local maxima of the
TIC representation that exceed a threshold are selected as initial seeds. Then, the peak area
is determined by using the distance of the seed mass spectrum to all neighbor mass spectra
as a measure of the peak’s coherence. The area is extended until the distance exceeds a given
threshold. No information about the expected peak shape is needed. The peak integration
is based on the sum of TICs of the peak area. An identification of the area’s average or apex
mass spectrum or the seed mass spectrum is again possible using the MetaboliteDB module.
To represent the similarities and differences between different chromatograms, bidirectional
best hits are used to find co-occurring peaks. These are located by using a distance that
exponentially penalizes differences in the first and second retention times of the peaks to be
compared. To avoid a full computation of all pairs of peaks, only those peaks within a defined
window of retention times based on the standard deviation of the exponential time penalty
function are evaluated.
86
14 Metabolomics
Will-be-set-by-IN-TECH

ChromA4D’s visualizations represent aligned chromatograms as color overlay images, similar


to those used in differential proteomics. This allows a direct visual comparison of signals
present in one sample, but not present in another sample.
ChromA4D creates peak report tables in CSV format, which include peak apex positions in
both chromatographic dimensions, area under curve, peak intensity and possibly tentative
database identifications. Additionally, information about the matched and aligned peak
groups is saved in CSV format.

5. Application examples
The following examples for GC-MS and GCxGC-MS are based on the Maltcms framework,
using the ChromA and ChromA4D configurations described in the previous sections. In order
to run them, the recent version of Maltcms needs to be downloaded and unzipped to a local
folder on a computer. Additionally, Maltcms requires a JAVA runtime environment version 6
or newer to be installed. If these requirements are met, one needs to start a command prompt
and change to the folder containing the unzipped Maltcms.

5.1 An example workflow for GC-MS


The experiment used to illustrate an example workflow for one-dimensional GC-MS consists
of two samples of standard compounds, which contain mainly sugars, amino acids,
other organic acids and nucleosides, measured after manual (MD) and after automatic
derivatization (AD) with the derivatization protocol and substances given below. Group
AD consists of a sample of n-alkanes standard and two replicates of mix1, namely mix1-1
and mix1-2. We will show how ChromA can be used to find and integrate peaks, as well as
compare and align the peaks between the samples, and finally how the alignment results can
be used for quality control.

5.1.1 Sample preparation


20 μL of each sample were incubated with 60 μL methoxylamine hydrochloride
(Sigma Aldrich) in pyridine (20 mg/ml) for 90 min at 60◦ C before 100 μL of N-
Methyl-N-(trimethylsilyl)-trifluoroacetamide (MSTFA) (Macherey & Nagel) were added for
60 min at 37◦ C.

5.1.2 Acquisition and data processing


The samples were acquired on an Agilent GC 7890N with MSD 5975C triple axis detector. An
Agilent HP5ms column with a length of 30 m, a diameter of 0.25 mm, and a film thickness
of 0.25 μm (Agilent, Santa Clara CA, USA) was used for the gas-chromatographic separation,
followed by a deactivated restriction capillary with 50 cm length and a diameter of 0.18 mm.
Per sample, 1 μL was injected onto the column in pulsed splitless mode (30 psi for 2 min).
The flow rate was set to 1.5 mL/min of Helium. The linear temperature ramp started at 50 ◦ C
for 2 min until it reached its maximum of 325 ◦ C at a rate of 10 ◦ C/min. The raw data were
exported to NetCDF format using the Agilent ChemStation software v.B.04.01 (Agilent, Santa
Clara CA, USA) with default parameters and without additional preprocessing applied.
A sample containing n-alkanes was measured as an external standard for manual (MD) and
automatic derivatization (AD) in order to be able to later determine retention indices for
Generic
GC-MS BasedSoftware
Metabolomics Frameworks for GC-MS Based Metabolomics 87
Generic Software Frameworks for
15

(a) Overlay of unaligned data sets, extracted from middle section within a time range of 1100 to 1700
seconds.

(b) Overlay with highlighted peak areas (without n-alkanes) after peak finding and integration. Zoomed
in to provide more detail.

Fig. 2. TIC overlay plots of the raw GC-MS data sets.


88
16 Metabolomics
Will-be-set-by-IN-TECH

the other samples. The acquired data were exported to ANDI-MS (NetCDF) format before
ChromA was applied. The default ChromA pipeline chroma.properties was run from the
unzipped Maltcms directory with the following command (issued on a single line of input):

> java -Xmx1G -jar maltcms.jar -i ../data/ -o ../output/ -f *.CDF \


-c cfg/chroma.properties

-i points to the directory containing the input data, -o points to the directory where output
should be placed, -f can be a comma separated list of filenames or, as in this case, a wildcard
expression, matching all files in the input directory having a file name ending with .CDF.
The final argument indicated by -c is the path to the configuration file used for definition
of the pipeline and its commands. An overlay of the raw TICs of the samples is depicted in
Figure 2(a). The default ChromA pipeline configuration creates a profile matrix with nominal
mass bin width. Then, the TIC peaks are located separately within each sample data file and
are integrated (Figure 2(b)). The peak apex mass spectra are then used in the next step in
order to build a multiple peak alignment between all peaks of all samples by finding large
cliques, or clusters of peaks exhibiting similar retention time behaviour and having highly
similar mass spectra. This coarse alignment could already be used to calculate a polynomial
fit, correcting retention time shift for all peaks. However, the ChromA pipeline uses the
peak clusters in order to constrain a dynamic time warping (DTW) alignment in the next
step, which is calculated between all pairs of samples. The resulting distances are used to
determine the reference sample with the lowest sum of distances to all remaining samples.
Those are then aligned to the reference using the warp map obtained from the pairwise
DTW calculations. The pairwise DTW distances can easily be used for a hierarchical cluster
analysis. Similar samples should be grouped into the same cluster, while dissimilar samples
should be grouped into different clusters. Figure 3 shows the results of applying a complete
linkage clustering algorithm provided by GNU R to the pairwise distance matrix. It is clearly
visible that the samples are grouped correctly, without incorporation of any external group
assignment. Thus, this method can be used for quality control of multiple sample acquisitions,
when the clustering results are compared against a pre-defined number of sample groups.

5.2 An example workflow for GCxGC-MS


The instructional samples presented in this section were preprocessed according to the
protocol given by Doebbe et al. (2010). The description of the protocol has been adapted
from that reference where necessary.

5.2.1 Sample preparation


The samples were incubated with 100 μl methoxylamine hydrochloride (Sigma
Aldrich) in pyridine (20 mg/ml) for 90 min at 37◦ C while stirring. N-
Methyl-N-(trimethylsilyl)-trifluoroacetamide (MSTFA) (Macherey & Nagel) was then
added and incubated for another 30 min at 37◦ C with constant stirring.

5.2.2 Acquisition and data processing


The sample acquisition was performed on a LECO Pegasus 4D TOF-MS (LECO, St. Joseph,
MI, USA). The Pegasus 4D system was equipped with an Agilent 6890 gas chromatograph
Generic
GC-MS BasedSoftware
Metabolomics Frameworks for GC-MS Based Metabolomics 89
Generic Software Frameworks for
17

Complete linkage clustering of pairwise DTW distances


0.35
0.30
0.25
0.20
0.15
0.10
Height

0.05
0.00

mix1−MD.cdf
n−alkanes−AD.cdf

n−alkanes−MD.cdf

mix1−1−AD.cdf

mix1−2−AD.cdf
as.dist(1 − tbl)

Fig. 3. Clustering of GC-MS samples based on pairwise DTW similarities transformed to


distances. The samples are clearly separated into two clusters, one containing the n-alkane
standard samples, the other one containing the mix1 samples.

(Agilent, Santa Clara, CA, USA). The inlet temperature was set to 275◦ C. An Rtx-5ms (Restek,
Bellefonte, PA, USA) capillary column was used with a length of 30 m, 0.25 mm diameter
and 0.25 μm film thickness as the primary column. The secondary column was a BPX-50
(SGE, Ringwood, Victoria, Australia) capillary column with a length of 2 m, a diameter of
0.1 mm and 0.1 μm film thickness. The temperature program of the primary oven was set to
the following conditions: 70◦ C for 2 min, 4◦ C/min to 180◦ C, 2◦ C/min to 230◦ C, 4◦ C/min
to 325◦ C hold 3 min. This program resulted in a total runtime of about 70 min for each
sample. The secondary oven was programmed with an offset of 15◦ C to the primary oven
temperature. The thermal modulator was set 30◦ C relative to the primary oven and to a
modulation time of 5 seconds with a hot pulse time of 0.4 seconds. The mass spectrometer ion
source temperature was set to 200◦ C and the ionization was performed at -70eV. The detector
voltage was set to 1600V and the stored mass range was 50-750 m/z with an acquisition rate
of 200 spectra/second.
90

(a) 2D-TIC plot before filters were applied. Long tailing peaks are (b) 2D-TIC plot after application of a moving median filter with
visible within the vertical dimension. Additionally, high frequency window size 3 for smoothing of high-frequency noise and successive
noise is present in the raw exported data, which is barely visible at application of a top hat filter with a window size of 301 for baseline
this resolution. removal in order to reduce false positive peak finding results.

Fig. 4. Visualizations of Standard-Mix1-1 before and after signal filtering with the ChromA4D processing pipeline.
Metabolomics
Generic
GC-MS BasedSoftware
Generic Software Frameworks for
Metabolomics Frameworks for GC-MS Based Metabolomics

(a) 2D-TIC plot of Standard-Mix1-1 after peak finding and (b) Differential plot of the two Standard-Mix1 samples after DTW
integration with seeded region growing based on the cosine mass alignment based on vertical TIC slices. Yellow color indicates
spectral similarity with a fusion threshold of 0.99. Peak areas were similar amounts of total ion intensity in both samples. Green
limited to contain at most 100 points. shows a surplus in Standard-Mix1-1, while red shows a surplus in
Standard-Mix1-2.

Fig. 5. Visualizations of Standard-Mix1-1 after peak finding and of Standard-Mix1-1 and Standard-Mix1-2 after alignment with DTW.
19
91
92
20 Metabolomics
Will-be-set-by-IN-TECH

The raw acquired samples in LECO’s proprietary ELU format were exported to NetCDF
format using the LECO ChromaTOF software v.4.22 (LECO, St. Joseph, MI, USA). Initial
attempts to export the full, raw data failed with a crash beyond a NetCDF file size of 4GBytes.
Thus, we resampled the data with ChromaTOF to 100 Hz (resampling factor 2) and exported
with automatic signal smoothing and baseline offset correction value of 1 which resulted
in file sizes around 3GBytes per sample. The samples presented in this section are named
"Standard-Mix1-1" and "Standard-Mix1-2" and were measured on different days (Nov. 29th,
2008 and Dec. 12th, 2008).
The default ChromA4D pipeline for peak finding was called from within the unzipped
Maltcms directory (issued on a single line of input):

> java -Xmx2G -jar maltcms.jar -i ../data/ -o ../output/ \


-f *.cdf -c cfg/4Dpeakfinding.properties

The pipeline first preprocesses the data by applying a median filter followed by a top hat
filter in order to remove high- and low-frequency noise contributions (Figures 4(a) and 4(b)).
ChromA4D then uses a variant of seeded region growing in order to extend peak seeds, which
are found as local maxima of the 2D-TIC. These initial seeds are then extended until the mass
spectral similarity of the seed and the next evaluated candidate drops below a user-defined
threshold, or until the peak area reaches its maximum, pre-defined size (Figure 5(a)). After
peak area integration, the pipeline clusters peaks between samples based on their mass
spectral similarity and retention time behaviour in both dimensions to form peak cliques
(not shown) as multiple peak alignments, which are then exported into CSV format for
further downstream processing. Another possible application shown in Figure 5(b) is the
visualization of pairwise GCxGC-MS alignments using DTW on the vertical 2D-TIC slices,
which can be useful for qualitative comparisons.

6. Summary and outlook


The present state of Open Source frameworks for metabolomics is very diverse. A number
of tools have seen steady development and improvement over the last years, such as
XCMS, MZmine, and PyMS, while others are still being developed, such as mzMatch,
Guineu, and Maltcms. There is currently no framework available that covers every aspect
of metabolomics data preprocessing. Most of the frameworks concentrate on one or a
few analytical technologies with the largest distinction being between GC-MS and LC-MS.
GCxGC-MS raw data processing is currently only handled by Maltcms’ ChromA4D pipeline,
while Guineu processes peak lists exported from LECO’s ChromaTOF software and offers
statistical methods for sample comparison together with a user-friendly graphical interface.
We showed two instructive examples on setting up and running the basic processing pipelines
ChromA and ChromA4D for GC-MS and GCxGC-MS raw data. The general structure of
these pipelines would be slightly different for each of the Open Source frameworks presented
in this chapter, however, the basic concepts behind the processing steps are the same for all
tools. Since metabolomics is an evolving field of research, no framework captures all possible
use-cases, but it will be interesting to see which frameworks will be flexible and extendable
enough to be adapted to new requirements in the near future.
Generic
GC-MS BasedSoftware
Metabolomics Frameworks for GC-MS Based Metabolomics 93
Generic Software Frameworks for
21

In order to combine experiments from multiple "omics" experiments, another level of


abstraction on top of local or web-service based tools for data processing, fusion, and
integration of metabolomics experiments is a necessary future requirement. Generic workflow
systems like Taverna (Hull et al., 2006) or Conveyor (Linke et al., 2011) offer integration
of such resources, augmented with graphical editors for point-and-click user interaction.
However, due to their generic nature these systems are far away from being as user-friendly
as applications designed for a specific data analysis task and require some expert knowledge
when assembling task-specific processing graphs.
One point that requires further attention is the definition and controlled evolution of peak
data formats for metabolomics, along with other formats for easier exchange of secondary
data between applications and frameworks. A first step in this direction has been taken
by Scheltema et al. (2011) by defining the PeakML format. However, it is important that
such formats are curated and evolved, possibly by a larger non-profit organization like the
HUPO within its proteomics standards initiative HUPO PSI. Primary data is already acessible
in a variety of different, defined formats, the most recent addition being mzML (Martens
et al., 2010) which is curated by the PSI. Such standardization attempts can however only be
successful and gain the required momentum if also the manufacturers of analytical machinery
support the formats with their proprietary software within a short time frame after the
specification and see a benefit in offering such functionality due to the expressed demand
of scientists working in the field as in case of NetCDF, mzData, or mzML.

7. Acknowledgements
We would like to thank Manuela Meyer at the Center for Biotechnology (CeBiTec), Bielefeld
University, Germany, for the preparation of the GC-MS samples used in this work.
We furthermore thank Denise Schöfbeck and Rainer Schumacher, Center for Analytical
Chemistry, IFA Tulln, University of Natural Resources and Life Sciences, Vienna, Austria,
for measuring those samples and for kindly providing us with the datasets. Furthermore, we
would like to express our gratitude to Anja Döbbe and Olaf Kruse, Algae BioTech Group at the
CeBiTec, for providing the GCxGC-MS samples and datasets. We finally thank our students
Mathias Wilhelm for his work on ChromA4D and Kai-Bernd Stadermann for his work on the
RMI-based remote execution framework.

8. References
Åberg, K., Alm, E. & Torgrip, R. (2009). The correspondence problem for metabonomics
datasets, Analytical and Bioanalytical Chemistry 394(1): 151–162.
Ahmad, I., Suits, F., Hoekman, B., Swertz, M. A., Byelas, H., Dijkstra, M., Hooft, R., Katsubo,
D., van Breukelen, B., Bischoff, R. & Horvatovich, P. (2011). A high-throughput
processing service for retention time alignment of complex proteomics and
metabolomics LC-MS data, Bioinformatics 27(8): 1176–1178.
Babushok, V. I., Linstrom, P. J., Reed, J. J., Zenkevich, I. G., Brown, R. L., Mallard, W. G. & Stein,
S. E. (2007). Development of a database of gas chromatographic retention properties
of organic compounds., Journal of Chromatography A 1157(1-2): 414–421.
Berk, M., Ebbels, T. & Montana, G. (2011). A statistical framework for biomarker discovery in
metabolomic time course data, Bioinformatics 27(14): 1979–1985.
94
22 Metabolomics
Will-be-set-by-IN-TECH

Bertsch, A., Hildebrandt, A., Hussong, R. & Zerck, A. (2008). OpenMS - An open-source
software framework for mass spectrometry., BMC Bioinformatics 9(1): 163.
Biller, J. E. & Biemann, K. (1974). Reconstructed Mass Spectra, A Novel Approach for
the Utilization of Gas Chromatograph—Mass Spectrometer Data, Analytical Letters
7(7): 515–528.
Callaghan, S., De Souza, D., Tull, D., Roessner, U., Bacic, A., McConville, M. & Likić, V. (2010).
Application and comparative study of PyMS Python toolkit for processing of gas
chromatography-mass spectrometry (GC-MS) data, 2nd Australasian Symposium on
Metabolomics, Melbourne 2010.
Carroll, A. J., Badger, M. R. & Harvey Millar, A. (2010). The MetabolomeExpress Project:
enabling web-based processing, analysis and transparent dissemination of GC/MS
metabolomics datasets, BMC Bioinformatics 11(1): 376.
Castillo, S., Gopalacharyulu, P., Yetukuri, L. & Orešič, M. (2011). Algorithms and tools for the
preprocessing of LC–MS metabolomics data, Chemometrics and Intelligent Laboratory
Systems 108(1): 23–32.
Castillo, S., Mattila, I., Miettinen, J., Orešič, M. & Hyötyläinen, T. (2011). Data Analysis Tool
for Comprehensive Two-Dimensional Gas Chromatography/Time-of-Flight Mass
Spectrometry, Analytical Chemistry 83(8): 3058–3067.
Chae, M., Reis, R. & Thaden, J. J. (2008). An iterative block-shifting approach to retention
time alignment that preserves the shape and area of gas chromatography-mass
spectrometry peaks, BMC Bioinformatics 9(Suppl 9): S15.
Christin, C., Hoefsloot, H. C. J., Smilde, A. K., Suits, F., Bischoff, R. & Horvatovich, P. L.
(2010). Time Alignment Algorithms Based on Selected Mass Traces for Complex
LC-MS Data, Journal of Proteome Research 9(3): 1483–1495.
Clifford, D., Stone, G., Montoliu, I., Rezzi, S., Martin, F.-P., Guy, P., Bruce, S. & Kochhar,
S. (2009). Alignment Using Variable Penalty Dynamic Time Warping, Analytical
Chemistry 81(3): 1000–1007.
Deutsch, E. (2008). mzML: a single, unifying data format for mass spectrometer output.,
Proteomics 8(14): 2776–2777.
Deutsch, E. W., Mendoza, L., Shteynberg, D., Farrah, T., Lam, H., Tasman, N., Sun, Z., Nilsson,
E., Pratt, B., Prazen, B., Eng, J. K., Martin, D. B., Nesvizhskii, A. I. & Aebersold, R.
(2010). A guided tour of the Trans-Proteomic Pipeline, Proteomics 10(6): 1150–1159.
Doebbe, A., Keck, M., Russa, M. L., Mussgnug, J. H., Hankamer, B., Tekce, E., Niehaus, K.
& Kruse, O. (2010). The Interplay of Proton, Electron, and Metabolite Supply for
Photosynthetic H2 Production in Chlamydomonas reinhardtii, Journal of Biological
Chemistry 285(39): 30247–30260.
Du, P., Kibbe, W. A. & Lin, S. M. (2006). Improved peak detection in mass spectrum by
incorporating continuous wavelet transform-based pattern matching, Bioinformatics
22(17): 2059–2065.
Foster, I. T. (2005). Globus Toolkit Version 4: Software for Service-Oriented Systems., in H. Jin,
D. A. Reed & W. Jiang (eds), IFIP International Conference on Network and Parallel
Computing, Springer, pp. 2–13.
Fredriksson, M. J., Petersson, P., Axelsson, B.-O. & Bylund, D. (2009). An automatic peak
finding method for LC-MS data using Gaussian second derivative filtering., Journal
of Separation Science 32(22): 3906–3918.
GNU R (2011).
URL: http://www.r-project.org/
Generic
GC-MS BasedSoftware
Metabolomics Frameworks for GC-MS Based Metabolomics 95
Generic Software Frameworks for
23

Hoffmann, N. & Stoye, J. (2009). ChromA: signal-based retention time alignment for
chromatography-mass spectrometry data, Bioinformatics 25(16): 2080–2081.
Hull, D., Wolstencroft, K., Stevens, R., Goble, C., Pocock, M. R., Li, P. & Oinn, T. (2006).
Taverna: a tool for building and running workflows of services, Nucleic Acids Research
34(suppl 2): W729–W732.
Hummel, J., Selbig, J., Walther, D. & Kopka, J. (2007). The Golm Metabolome Database:
a database for GC-MS based metabolite profiling, in J. Nielsen & M. Jewett (eds),
Metabolomics, Springer Berlin / Heidelberg, pp. 75–95.
HUPO PSI (2011).
URL: http://www.psidev.info/
Isaac, A., Lee, L., Keen, W., Erwin, T., Wang, Q., De Souza, D., Roessner, U., Pyke, J., Kotagiri,
R., Wettenhall, R., McConville, M., Bacic, A. & Likić, V. (2009). PyMS: A Python
toolkit for processing of gas chromatography-mass spectrometry data, Bioinformatics
Australia Conference, Melbourne 2009.
JAVA (2011).
URL: http://www.java.com/en/
Kankainen, M., Gopalacharyulu, P., Holm, L. & Orešič, M. (2011). MPEA–metabolite pathway
enrichment analysis, Bioinformatics 27(13): 1878–1879.
Kastenmüller, G., Römisch-Margl, W., Wägele, B., Altmaier, E. & Suhre, K. (2011).
metaP-Server: A Web-Based Metabolomics Data Analysis Tool, Journal of Biomedicine
and Biotechnology 2011: 1–8.
Kessner, D., Chambers, M., Burke, R., Agus, D. & Mallick, P. (2008). ProteoWizard:
open source software for rapid proteomics tools development., Bioinformatics
24(21): 2534–2536.
Kim, S., Fang, A., Wang, B., Jeong, J. & Zhang, X. (2011). An Optimal Peak Alignment For
Comprehensive Two-Dimensional Gas Chromatography Mass Spectrometry Using
Mixture Similarity Measure, Bioinformatics 27(12): 1660–1666.
Krebs, M. D., Tingley, R. D., Zeskind, J. E., Holmboe, M. E., Kang, J.-M. & Davis, C. E.
(2006). Alignment of gas chromatography-mass spectrometry data by landmark
selection from complex chemical mixtures, Chemometrics and Intelligent Laboratory
Systems 81(1): 74–81.
Lange, E., Gropl, C., Schulz-Trieglaff, O., Huber, C. & Reinert, K. (2007). A geometric approach
for the alignment of liquid chromatography mass spectrometry data, Bioinformatics
23(13): i273–i281.
Linke, B., Giegerich, R. & Goesmann, A. (2011). Conveyor: a workflow engine for
bioinformatic analyses, Bioinformatics 27(7): 903–911.
Lommen, A. (2009). MetAlign: Interface-Driven, Versatile Metabolomics Tool for
Hyphenated Full-Scan Mass Spectrometry Data Preprocessing, Analytical Chemistry
81(8): 3079–3086.
Maltcms (2011).
URL: http://maltcms.sourceforge.net
Martens, L., Chambers, M., Sturm, M., Kessner, D., Levander, F., Shofstahl, J., Tang,
W. H., Rompp, A., Neumann, S., Pizarro, A. D., Montecchi-Palazzi, L., Tasman, N.,
Coleman, M., Reisinger, F., Souda, P., Hermjakob, H., Binz, P. A. & Deutsch, E. W.
(2010). mzML–a Community Standard for Mass Spectrometry Data, Molecular and
Cellular Proteomics 10(1): R110.000133–R110.000133.
Matthews, L. (2000). ASTM Protocols for Analytical Data Interchange, 5(5): 60–61.
96
24 Metabolomics
Will-be-set-by-IN-TECH

Miura, D., Tsuji, Y., Takahashi, K., Wariishi, H. & Saito, K. (2010). A strategy
for the determination of the elemental composition by fourier transform ion
cyclotron resonance mass spectrometry based on isotopic peak ratios., Technical
Report 13, Innovation Center for Medical Redox Navigation, Kyushu University, 3-1-1
Maidashi, Higashi-ku, Fukuoka 12-8582, Japan.
Neumann, S. & Böcker, S. (2010). Computational mass spectrometry for metabolomics:
Identification of metabolites and small molecules, Analytical and Bioanalytical
Chemistry 398(7-8): 2779–2788.
Neuweger, H., Albaum, S. P., Niehaus, K., Stoye, J. & Goesmann, A. (2008). MeltDB: a
software platform for the analysis and integration of metabolomics experiment data,
Bioinformatics 24(23): 2726–2732.
Neuweger, H., Persicke, M., Albaum, S. P., Bekel, T., Dondrup, M., Hüser, A. T., Winnebald,
J., Schneider, J., Kalinowski, J. & Goesmann, A. (2009). Visualizing post genomics
data-sets on customized pathway maps by ProMeTra-aeration-dependent gene
expression and metabolism of Corynebacterium glutamicum as an example., BMC
Systems Biology 3: 82.
Oh, C., Huang, X., Regnier, F. E., Buck, C. & Zhang, X. (2008). Comprehensive
two-dimensional gas chromatography/time-of-flight mass spectrometry peak
sorting algorithm, Journal of Chromatography A 1179(2): 205–215.
Oliver, S. G., Paton, N. W. & Taylor, C. F. (2004). A common open representation of mass
spectrometry data and its application to proteomics research, Nature Biotechnology
22(11): 1459–1466. 10.1038/nbt1031.
Orchard, S., Hermjakob, H., Taylor, C. F., Potthast, F., Jones, P., Zhu, W., Julian, R. K. &
Apweiler, R. (2005). Second proteomics standards initiative spring workshop., Expert
review of proteomics, EMBL Outstation - European Bioinformatics Institute, Wellcome
Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK. pp. 287–289.
Pierce, K. M., Hoggard, J. C., Hope, J. L., Rainey, P. M., Hoofnagle, A. N., Jack, R. M.,
Wright, B. W. & Synovec, R. E. (2006). Fisher Ratio Method Applied to Third-Order
Separation Data To Identify Significant Chemical Components of Metabolite Extracts,
Analytical Chemistry 78(14): 5068–5075.
Pierce, K. M., Wood, L. F., Wright, B. W. & Synovec, R. E. (2005). A comprehensive
two-dimensional retention time alignment algorithm to enhance chemometric
analysis of comprehensive two-dimensional separation data, Analytical Chemistry
77(23): 7735–7743.
Pluskal, T., Castillo, S., Villar-Briones, A. & Orešič, M. (2010). MZmine 2: Modular
framework for processing, visualizing, and analyzing mass spectrometry-based
molecular profile data, BMC Bioinformatics 11(1): 395.
Prince, J. & Marcotte, E. (2006). Chromatographic alignment of ESI-LC-MS proteomics
data sets by ordered bijective interpolated warping, Analytical Chemistry
78(17): 6140–6152.
Python (2008).
URL: http://www.python.org/download/releases/2.5.2/
Rew, R. & Davis, G. (1990). NetCDF: an interface for scientific data access, Computer Graphics
and Applications, IEEE 10(4): 76–82.
Robinson, M. D., De Souza, D. P., Saunders, E. C., Mcconville, M. J., Speed, T. P. & Likić,
V. A. (2007). A dynamic programming approach for the alignment of signal peaks in
Generic
GC-MS BasedSoftware
Metabolomics Frameworks for GC-MS Based Metabolomics 97
Generic Software Frameworks for
25

multiple gas chromatography-mass spectrometry experiments, BMC Bioinformatics


8(1): 419.
Savitzky, A. & Golay, M. J. E. (1964). Smoothing and Differentiation of Data by Simplified
Least Squares Procedures., Analytical Chemistry 36(8): 1627–1639.
Scheltema, R. A., Jankevics, A., Jansen, R. C., Swertz, M. A. & Breitling, R. (2011).
PeakML/mzMatch: A File Format, Java Library, R Library, and Tool-Chain for Mass
Spectrometry Data Analysis, Analytical Chemistry 83(7): 2786–2793.
SciPy (2011).
URL: http://www.scipy.org/
Smilde, A. K. & Horvatovich, P. L. (2008). Optimized time alignment algorithm for LC-MS
data: correlation optimized warping using component detection algorithm-selected
mass chromatograms, Analytical Chemistry 80(18): 7012–7021.
Smith, C. A., Want, E. J., O’Maille, G., Abagyan, R. & Siuzdak, G. (2006). XCMS: Processing
Mass Spectrometry Data for Metabolite Profiling Using Nonlinear Peak Alignment,
Matching, and Identification, Analytical Chemistry 78(3): 779–787.
Stein, S. (1999). An integrated method for spectrum extraction and compound identification
from gas chromatography/mass spectrometry data, Journal of the American Society for
Mass Spectrometry 10(8): 770–781.
Tautenhahn, R., Böttcher, C. & Neumann, S. (2007). Annotation of LC/ESI-MS Mass Signals,
in S. Hochreiter & R. Wagner (eds), Bioinformatics Research and Development, Springer
Berlin / Heidelberg, pp. 371–380. 10.1007/978-3-540-71233-6_29.
Tautenhahn, R., Böttcher, C. & Neumann, S. (2008). Highly sensitive feature detection for high
resolution LC/MS, BMC Bioinformatics 9: 504.
Tohge, T. & Fernie, A. R. (2009). Web-based resources for mass-spectrometry-based
metabolomics: A user’s guide, Phytochemistry 70(4): 450–456.
Vial, J., Noçairi, H., Sassiat, P., Mallipatu, S., Cognon, G., Thiébaut, D., Teillet, B. & Rutledge,
D. N. (2009). Combination of dynamic time warping and multivariate analysis for
the comparison of comprehensive two-dimensional gas chromatograms: application
to plant extracts, Journal of Chromatography A 1216(14): 2866–2872.
Wang, S. Y., Ho, T. J., Kuo, C. H. & Tseng, Y. J. (2010). Chromaligner: a web server for
chromatogram alignment, Bioinformatics 26(18): 2338–2339.
Wiklund, S., Johansson, E., Sjöström, L., Mellerowicz, E. J., Edlund, U., Shockcor, J. P.,
Gottfries, J., Moritz, T. & Trygg, J. (2008). Visualization of GC/TOF-MS-based
metabolomics data for identification of biochemically interesting compounds using
OPLS class models, Analytical Chemistry 80(1): 115–122.
Wishart, D. S., Knox, C., Guo, A. C., Eisner, R., Young, N., Gautam, B., Hau, D. D., Psychogios,
N., Dong, E., Bouatra, S., Mandal, R., Sinelnikov, I., Xia, J., Jia, L., Cruz, J. A., Lim,
E., Sobsey, C. A., Shrivastava, S., Huang, P., Liu, P., Fang, L., Peng, J., Fradette, R.,
Cheng, D., Tzur, D., Clements, M., Lewis, A., De Souza, A., Zuniga, A., Dawe, M.,
Xiong, Y., Clive, D., Greiner, R., Nazyrova, A., Shaykhutdinov, R., Li, L., Vogel, H. J.
& Forsythe, I. (2009). HMDB: a knowledgebase for the human metabolome, Nucleic
Acids Research 37(Database): D603–D610.
Xia, J., Sinelnikov, I. V. & Wishart, D. S. (2011). MetATT: a web-based metabolomics tool for
analyzing time-series and two-factor data sets, Bioinformatics 27(17): 2455–2456.
Xia, J. & Wishart, D. S. (2010a). MetPA: a web-based metabolomics tool for pathway analysis
and visualization, Bioinformatics 26(18): 2342–2344.
98
26 Metabolomics
Will-be-set-by-IN-TECH

Xia, J. & Wishart, D. S. (2010b). MSEA: a web-based tool to identify biologically


meaningful patterns in quantitative metabolomic data, Nucleic Acids Research
38(suppl 2): W71–W77.
Xia, J. & Wishart, D. S. (2011). Web-based inference of biological patterns, functions
and pathways from metabolomic data using MetaboAnalyst, Nature Protocols
6(6): 743–760.
XML (2008).
URL: http://www.w3.org/TR/REC-xml/
Zhang, X. (2010). DISCO: distance and spectrum correlation optimization alignment
for two-dimensional gas chromatography time-of-flight mass spectrometry-based
metabolomics, Analytical Chemistry 82(12): 5069–5081.
5

Computational Methods to Interpret and


Integrate Metabolomic Data
Feng Li1, Jiangxin Wang2, Lei Nie3 and Weiwen Zhang2
1Department of Mathematics and Statistics, University of Maryland,
Baltimore County, Baltimore, MD,
2School of Chemical Engineering and Technology, Tianjin University, Tianjin,
3Division of Biostatistics, Department of Epidemiology and Preventive Medicine,

University of Maryland Baltimore, Baltimore, MD,


2P.R. China
1,3U.S.A.

1. Introduction
Revolutionary improvements in high-throughput DNA sequencing technologies have made
it possible to measure gene, mRNA, proteins and metabolites, as well as their interaction at
global level. In the past decades, significant efforts in improving analytical technologies
pertaining to measuring mRNA, proteins and metabolites have been made. These efforts
have led to the generation of several new ‘omics’ research fields: transcriptomics,
proteomics, metabolomics, interactomics and so on (Singh & Nagaraj, 2006; Fiehn 2007; Lin
& Qian, 2007; Kandpal et al., 2009; Ishii & Tomita, 2009). Among them, metabolomics is an
approach to obtain a comprehensive evaluation of metabolites in cells. Compared with
transcriptomics and proteomics approaches, metabolomics can achieve large-scale
quantitative and qualitative measurements of cellular metabolites, which can thus generate
a high-resolution biochemical and functional information of an organism.
Due to the chemical complexity of cellular metabolites, it is generally accepted that no single
analytical technique can provide a comprehensive visualization of all metabolites, so
multiple technologies are generally employed (Dunn & Ellis, 2005; Villas-Boas Silas et al.,
2005; Hollywood et al., 2006; Dettmer et al., 2007; Lenz & Wilson, 2007; Seger & Sturm,
2007). The selection of the most suitable technology is typically a compromise between
speed, chemical selectivity, and instrumental sensitivity. Tools such as nuclear magnetic
resonance spectroscopy (NMR) are rapid, highly selective and non-destructive, but have
relatively lower sensitivities. Other tools such as capillary electrophoresis (CE) coupled to
laser-induced fluorescence detection are highly sensitive, but have limited chemical
selectivity (Ramautar et al., 2006). So far mass spectrometry (MS) measurement following
chromatographic separation offers the best combination of sensitivity and selectivity (Dunn
& Ellis, 2005; Bedair & Sumner, 2008). Mass-selective detection provides highly specific
chemical information including molecular mass and/or characteristic fragment-ion that is
directly related to chemical structure of molecules. This information can be utilized for
compound identification through spectral matching with data compiled in libraries for
100 Metabolomics

authentic compounds or used for de novo structural elucidation. Further, chemically selective
MS information can be obtained from extremely small quantities of metabolites in the pmole
and fmole level for many primary and secondary metabolites. Different technologies, either
individual or integrated, could be employed for different study aims, based on metabolite
identification, detection speed, high throughput and sensitivity. In the chapter, we will first
review current MS technologies that have been incorporated into many metabolomics
research programs as well as some of the emerging MS technologies that hold additional
promise for the future advancement of metabolomics.
In contrast to classical biochemical approaches that typically focus on a single metabolite,
single metabolic reaction or their kinetic properties, metabolomics involves collection of
large amount of quantitative data on a broad series of metabolites in an attempt to gain an
overall understanding of metabolism and/or metabolic dynamics associated with
conditions of interest, such as disease or drug exposure. Generally speaking, metabolomics
data share a great deal of similarity with transcriptomics data: both types of data matrices
are large, feature rich, and challenged with issues of dealing with a limited sample size and
a high-dimensional feature space. Thus in many cases the robust data processing algorithms
originally developed for transcriptomic analysis have been adapted directly for
metabolomic analysis. However, the challenges with metabolomic data can be unique, and
may require new methodologies supported with a detailed knowledge of cheminformatics,
bioinformatics, optimization, dynamic system theory and statistics. In recent years, many
computational methods have been developed specifically for metabolomic data, varying
from metabolic network analysis oriented or feature selection/data mining oriented. In the
chapter, we will introduce each of these methods and their relevant applications, and will
also discuss all the computational challenges associated.

2. Major analytical technologies of metabolomics


2.1 Mass spectrometry (MS)-based metabolomics
Several MS-based metabolomics technologies have been developed in the past decades, a
brief introduction was presented here. Much detailed information regarding these
technologies can be found from several recent excellent reviews (Dunn & Ellis, 2005;
Hollywood et al., 2006; Dettmer et al., 2007; Bedair & Sumner, 2008).
Direct infusion mass spectrometry (DIMS) is a method for direct analysis of complex
metabolic extracts without extraction or separation via electrospray ionization (ESI) MS,
which provides a sensitive, high-throughput method to make it possible for several hundred
samples per day. In terms of disadvantages, DIMS analysis is susceptible to ionization
suppression due to competitive ionization with other components in the matrix (e.g., salts
and other ionic compounds, organic acids/bases, and hydrophobic compounds), although
ionization suppression effects could be reduced by nano electrospray (nano-DIMS) with
increased ionization efficiency (Southam et al., 2007). In addition, typical DIMS is not able to
discriminate isomeric compounds; however, when coupled with tandem MS (DIMSMS) or
Fourier transform ion cyclotron resonance (FT-ICR) spectrometers can trap and accumulate
fragment ions that often enable to determine different isomeric structures (Aharoni et al.,
2002). A comparison between direct infusion negative-ESI iontrap MS and GC-quadrupole
MS analysis for the metabolic fingerprinting of five yeast mutants was reported recently
Computational Methods to Interpret and Integrate Metabolomic Data 101

(Mas et al., 2007). Negative ESI LCQ ion-trap MS was reported as an effective method for the
characterization of plant extracts with well-defined clusters in comparison to positive-ion
ESI and 1H-NMR profiling (Mattoli et al., 2006).
Gas chromatography-mass spectrometry (GC-MS) has been a very useful technology for
volatile and thermally stable polar and nonpolar metabolites (Tanaka et al., 1980).
Metabolite identification or confirmation is performed by retention time or index
comparisons with pure compounds and mass spectral interpretation or comparison using
retention index/mass spectral library databases (Wagner et al., 2003). Metabolites can be
classified into two classes: volatile metabolites not requiring chemical derivatisation
(Yassaa et al., 2001; Mallouchos et al., 2002; Deng et al., 2004) and non-volatile metabolites
requiring chemical derivatisation (Roessner et al., 2000). GC-MS based metabolic profiling
has been used to compare four Arabidopsis genotypes and showed each genotype
exhibited a different metabolite profile (Birkemeyer et al., 2003), and to compare
transgenic tomato plants over expressing hexokinase (Roessner-Tunali et al., 2003). Silent
phenotypes of potatoes have been distinguished from their parental background by
employing metabolic profiling (Weckwerth et al., 2004). The same approach has recently
been employed in microbial metabolomics to study the effect of different growth
conditions on Corynebacterium glutamicum (Strelkov et al., 2004).
The application of Liquid Chromatography coupled to Mass Spectrometry (LC-MS) in
metabolomics has been growing over the past few years (Wittmann et al., 2004). As a
universal separation technique that can be tailored for the targeted analysis of specific
metabolite groups or utilized in a broader non-targeted manner, LC offers additional
benefits of analyte recovery by fraction collection and/or concentration, which has been
difficult for GC separation. In addition, LC-MS operates at lower analysis temperatures than
GC-MS, which enables the analysis of heatlabile metabolites. LC-MS analysis does not
involve sample derivatization, which simplifies the sample-preparation and improves the
identification of metabolites. The major disadvantage of LC-MS relative to GC-MS is the lack
of transferable LC-MS libraries for metabolite identifications, although some efforts have
been initiated to construct in-house LC-MS or LC-MS-MS libraries for automated metabolite
identifications (Noteborn et al., 2000). Two-dimensional LC has also been utilized to increase
the peak separation capacity (Aharoni et al., 2002). Recent LC-MS metabolite-profiling
examples include the identification of flavonoids and isoflavonoids in Medicago truncatula
(Daykin et al., 2002), the revelation of novel pathways by studying the differential and
elicitor-specific responses in phenylpropanoid and isoflavonoid biosynthesis in Medicago
truncatula cell cultures (Farag et al., 2008), and the investigation of small polar-metabolite
responses to salt stress in Arabidopsis thaliana (Lindon et al., 2000). LC-MS has also been used
in the non-targeted analysis of endogenous metabolites in an unbiased manner (Rashed et
al., 1997; De Vos et al., 2007).
Capillary electrophoresis mass spectrometry (CE-MS) is a powerful separation technique for
charged metabolites (Ramautar et al., 2006; Monton et al., 2007). CE has superior separation
efficiencies compared to LC due to the plug-flow profile generated by the electroosmotic
flow (EOF) as compared to the parabolic flow in LC. Capillary zone electrophoresis (CZE)
has been the major CE mode used for CE-MS analysis of metabolites, due to the simplicity of
the running buffer. Simultaneous separation of charged and neutral metabolites can be
achieved using other CE modes (e.g., micellar electrokinetic chromatography (MEKC) or
102 Metabolomics

capillary electrochromatography (CEC)). Cationic and anionic CE-MS analysis of Bacillus


subtilis extracts detected 1692 metabolite features of which 150 were identified (Soga et al.,
2003). The same analytical procedure was recently used to study the alteration of metabolic
pathways in transgenic rice lines that over-express dihydroflavonol-4-reductase (Sato et al.,
2004). Non-aqueous CE-ESI ion-trap MSn was utilized for quantitative and qualitative
profiling of isoquinoline alkaloids in single-plant tubers of four central European Corydalis
species (Sturm et al., 2007).

2.2 Other emerging mass spectrometry technologies


Matrix assisted laser desorption ionization mass spectrometry (MALDI-MS) is a popular
analytical technique for biopolymer analysis. It has a high throughput capacity and a higher
tolerance for salts than ESI. In metabolomics, MALDI has largely been confined to the
targeted analysis of high-molecular-weight metabolites due to the substantial chemical-
background signals generated by the matrix in the low-molecular-weight region (<1,000
m/z) (e.g., the analysis of phospholipids in mammalian tissues (Jones et al., 2006), plant
carotenoids (Fraser et al., 2007), and plant cell wall xyloglycans (Lerouxel et al., 2002).
MALDI has also been used for imaging MS (IMS) of proteins and small molecules in tissues
(Reyzer & Caprioli, 2007). Whole organisms or selected tissue sections are analyzed through
an array of spots in which MS spectra are acquired at spatial intervals that define the image
resolution. The m/z intensities of the acquired spectra are then plotted in the x and y
coordinates to form a 2D image of the m/z values, which represents the spatial distribution
of that metabolite/ion in the tissue (Rubakhin et al., 2005). MALDI-TOF IMS has been
applied successfully for the study of drug and metabolite distributions in rat-brain tissues
(Hsieh et al., 2006) and whole rat body (Khatib-Shahidi et al., 2006).
Desorption electrospray ionization (DESI) is a new, ambient, soft-ionization technique that
combines features from both ESI and desorption ionization (DI) methods (Takats et al, 2004;
Cooks et al., 2006). In DESI, an electrospray emitter is used to generate a spray of charged
micro droplets that is directed towards an ambient sample surface. There is virtually no
sample preparation required for DESI, thus allowing the direct analysis of animal and plant
tissues. The application of DESI in metabolomics is relatively new, but its ambient DI
properties as well as its high-throughput capacity make it an attractive tool for
metabolomics. One promising area for DESI is in vivo metabolomics, which was
demonstrated through the direct profiling of alkaloids from plant tissues of Conium
maculatum without sample preparation while still identifying all of its previously reported
alkaloids using tandem MS (Talaty et al., 2005). Although this technique can be incorporated
into an IMS configuration, the spatial resolution of the DESI source (0.5–1.0 mm) is currently
less than that of MALDI ion imaging (50–100 μm) (Wiseman et al., 2005).
Extractive electrospray ionization (EESI) is another new ESI technique that uses two
separate sprayers. Although the exact sample-ionization mechanism is still unclear, the
ionization process depends on liquid–liquid extraction between the colliding micro-droplets
of the sample spray and the charged reagent-solvent spray (Chen et al., 2006). The
advantage of EESI is its ability to analyze complex biological samples, such as urine and
serum, directly with minimum or no sample preparation for an extended period of time.
EESI-MS along with 1H NMR was recently used to monitor the effect of diet on the
metabolites founds in rate urine (Gu et al., 2007).
Computational Methods to Interpret and Integrate Metabolomic Data 103

2.3 Nuclear magnetic resonance (NMR) spectroscopy


NMR spectroscopy is a technique that exploits the magnetic properties of certain atomic
nuclei to determine physical and chemical properties of atoms or the molecules in which
they are contained. It relies on the phenomenon of nuclear magnetic resonance and can
provide detailed information about the structure, dynamics, reaction state, and chemical
environment of molecules. Most frequently, NMR spectroscopy is used to investigate the
properties of organic molecules, though it is applicable to any nucleus possessing spin.
This can range from small compounds analyzed with 1-dimensional proton or carbon-13
NMR to large proteins or nucleic acids using 3 or 4-dimensional techniques (Grivet &
Delort, 2009). NMR presents an unbiased technique for metabolite fingerprinting that is
quantitative even in complex mixtures. Nicholson and co-workers have pioneered the
application of NMR for metabolite fingerprinting (Lindon et al., 2003). The analysis of six
yeast knock-out strains proved to classify and relate the genotypes by multivariate
statistics, which potentially can be applied for functional genomics (Raamsdonk et al.,
2001). NMR is non-destructive and therefore in vivo analysis is also possible (Gmati et al.,
2005). Thus, NMR provides a powerful method for accessing metabolite complement
(metabolome) and metabolic fluxes (fluxome) at a fine scale (metabolite identification)
and a global scale (metabolomics).

2.4 Vibrational spectroscopy


Vibrational spectroscopy is one of the oldest spectroscopic methods. The vibrational states
of a molecule can be probed in a variety of ways. The most direct way is through infrared
spectroscopy (IRS), as vibrational transitions typically require an amount of energy that
corresponds to the infrared region of the spectrum. Raman spectroscopy (RS), which
typically uses visible light, can also be used to measure vibration frequencies directly. RS
has been used for the identification of microorganisms of medical relevance (Dunn et al.,
2005); however, its application for complex biological systems outside the area of
microbiology is still in its infancy, although the potential of using 1064 nm excitation has
been demonstrated in studies of the biochemical analysis of honey (de Oliveira et al., 2002)
and in the analysis of plant pigments and essential oils (Schrader et al., 1999). In contrast,
IRS has been applied for diagnostics, characterisation of microorganism and plant,
adulteration and quality assurance, biomarker discovery and biochemical responses (Dunn
et al., 2005).

2.5 Single-cell metabolomics


Single cell analysis is the new frontier in “OMICS” (Wang & Bodoritz, 2010). Most current
metabolomic technologies only collect data averaged over thousands or millions of cells.
However, cellular heterogeneity within a cell population is a widespread event (Irish et al.,
2006; Graf & Stadtfeld, 2008). Since the metabolome provides biological processes occurring
in the cells, it will be imperative to establish a reliable metabolomic method to measure at a
single-cell level.
The challenges for single-cell metabolomics include: i) tiny quantities of metabolites from a
single cell. A type single cell is about 1-500 fL in volume (from E. coli to large mammalian),
104 Metabolomics

and with metabolites as low as amole to fmole (Schmid A et al., 2010), 106 times lower than a
typical population-based metabolomics. While amplification of DNA/RNA and highly
sensitive fluorescence measurements could be employed in single cell genomics,
transcriptomics and proteomics, no similar technique is available for single-cell
metabolomics. ii) Sample processing for a single cell is extremely challenging. Even though
detection limits for metabolites using MS can be as low as fmoles to amole range
(Amantonico et al., 2008); however, transferring of a cell or cell content to mass
spectrometer, conserving the original metabolome, and separating metabolites from cell
debris, proteins and salts, would be critical.
In recent years, several approaches have been established for MS-based single-cell
metabolomics (Figure 1) (see review by Heinemann & Zenobi, 2011). i) Sampling the cell
contents with a micropipette, followed by injection into a mass spectrometer using a
nano-electrospray ionization (nano-ESI) source (Masujima 2009). This approach, probably
only suitable for very large cells, can only measure a few cells per hour; ii) Sample
preparation on a microfluidic chip, followed by deposition on a sample plate for (matrix-
assisted) laser desorption/ionization (MALDI or LDI) mass spectrometry (Lu et al., 2006;
Mellors et al., 2008; Amantonico et al., 2008, 2010; Holmes et al., 2009). Once a complete
setup is realized, it has the potential to generate high throughput data in an automated
way; iii) Cell arraying, single cells are deposited on a sample plate for LDI or MALDI
covered by a solvent-repelling, application of a MALDI matrix in an organic solvent will
then lyse the cells and extract the compounds of interest for analysis by MALDI. This
approach is a true high-throughput operation because the sample arraying can be
automated, and thus the speed of MS instrument is the only limited factor (1000s of
cells/hour) (Urban et al., 2010); iv) Imaging mass spectrometry, many modern mass
spectrometers have imaging capabilities, with a spatial resolution of typically ~50 µm
(MALDI or LDI), and ~1 µm (secondary ion mass spectrometry, SIMS), at relatively fast
acquisition speed (Fletcher, 2009). With SIMS, the distribution of ions such as Na+, K+,
Ca2+, as well as cationized cholesterol, lipids present at cell surfaces can be imaged
(Fletcher 2009). However, so far the data generated through this approach has been less
quantitative (Heinemann & Zenobi, 2011).

Fig. 1. Schematics of the four MS-based approaches for single cell metabolomics.
(Heinemann & Zenobi, 2011)
Computational Methods to Interpret and Integrate Metabolomic Data 105

Only very few metabolites can be analyzed directly in single cells by autofluorescence
(Amantonico et al., 2010); however, by incorporating fluorescent tags or probes, researchers
have been able to detect more metabolites. For example, Fehr et al. (2002) developed a
protein based nanosensor for detection of maltose uptake by living yeast cells. In another
study, a genetically encoded fluorescent sensor was expressed in living cells for detecting
adenylate nucleotides (Berg et al., 2009). However, it is still arguable whether the formation
of these foreign complexes between sensor and metabolites in cells will cause damages and
lead to alteration of physiological status of cells.
The high sensitivity of electrochemical detectors to electroactive species makes them suitable
for targeted studies of metabolites in single-cell analysis. A range of microscale
electrochemical methods have been introduced to monitor various physiological processes
(Huang & Kennedy, 1995). For example, release of metabolites, such as catecholamines and
oxygen, can be readily measured electrochemically (Cannon et al., 2000). Specific methods
targeting particular metabolic pathways in single cells have been in use for a long time,
including autoradiography of cells preincubated with radioisotopically labeled compounds
(Fliermans & Schmidt, 1975).
Other methods used in single cells including single-cell spectroscopy in conjunction with
image analysis for glycogen metabolism in yeast cells (Cahill et al., 2000), enzyme-catalyzed
luminescence method for dopamine release from a mammalian nerve cell (Shinohara &
Wang, 2007), synchrotron Fourier transform infrared spectromicroscopy for ethanol
formation in single living cells of unicellular algae (Goff et al., 2009), raman spectroscopy
(Schuster et al., 2000; Buckmaster et al., 2009; Hermelink et al., 2009) for detecting nucleic
bases and amino acids in single cells, and nuclear magnetic resonance (NMR) for structural
characterization of organic compounds, including metabolites (Beckonert et al., 2007; Motta
et al., 2010). Brief summary and comparison for various metabolomics techniques discussed
above are listed in Table 1.

3. Computational methods for analyzing metabolomic data


Several computational methods have been developed in recent years to analyze
metabolomic data. The overview of the metabolomic data processing is shown in Figure 2.
These computational methods can be divided into two major categories: methods for data
pre-processing (low-level, such as noise reduction) or methods for interpretation (high-level,
such as feature selection). Pre-processing methods concern the improvement and the
enhancement of raw signals, which typically include noise reduction, peak detection,
baseline correction, peak alignment and normalization. Pre-processing methods for
metabolomic data has been reviewed in details by researchers from different perspectives
(Jewett et al., 2007; Enot et al., 2011). In this chapter, we will focus on the most widely used
or recently developed high-level methods for interpreting metabolomic data. Some
computational methods may also depend on the platform or instruments used. The reader
should refer Section 2 of the chapter for detailed explanation on popular platforms such as
GC-MS, LC-MS and NMR etc. Some software packages have been developed for
interpreting metabolomic data in recent years, although review of the software tools are
beyond the scope of this chapter, most of the software utilize the statistical methods
discussed here.
106 Metabolomics

Techniques Advantages Disadvatanges

nondiscrimination of isomeric
high-throughput, simple sample
DIMS compounds, susceptilble to
preparation
ionization

high sensitivity, ideal to complex derivation needed for non-volatile


GC-MS
samples, versatile metabolites

high sensitivity, average/high limited structural information, matrix


LC-MS
resolution effects

high sensitivity, quantification, label


CE-MS no robust, destructive
free, small volume

high sensitivity, detection of a wide


MALDI-MS no quantification, destructive
range of molecule, label free

direct analysis, high-throughput, optimization required for each


DESI
ambient depsorption ionization sample, low resolution

high-throughput, simple sample


EESI destructive
preparation

structural information, qualitative and expensive, time consuming, difficult


NMR
quantitative, versatile interpration

versatile, easy to identify functional poor structural information, difficult


IR
groups sample preparation, destructive

low selectivity, poor sturctural


Raman label free
information

high sensitivity, imaging capabilities, no structural information, targeted


FM
dynamic analysis only, difficult labelling

not comprehensive, no structural


high sensitivity, quantification, label
EC information, vulnerable to
free
interferences

FM: fluorescence microscopy


EC: electrochemical
Table 1. Summary for metabolomics techniques: characteristic of the main techniques
considered for application in metabolomics
Computational Methods to Interpret and Integrate Metabolomic Data 107

Fig. 2. An overview of metabolomic processing pipeline.

3.1 Computational methods for interpreting metabolomics


The computational methods to interpreting metabolomics data should be selected according
to the aim of a study. If the aim is for sample classification and prior information about the
sample identity is unknown, unsupervised methods such as hierarchical clustering analysis
(HCA), principal component analysis (PCA), or self-organizing map (SOM) are typically
used. On the other hand, in the case that sample identity is known and the aim of a study is
to discover characteristic biomarkers (e.g., search for biomarkers of a disease comparing
samples from healthy and diseased subjects), supervised methods such as linear
discriminant analysis, artificial neural networks (ANN) or support vector machine (SVM)
can be used. The supervised methods use prior information about sample class and perform
better in biomarker discovery. However, if the aim of the study is a mere biomarker
discovery in samples of known classes, common statistical methods such as ANOVA with
multiplicity control can also be used (Jonsson et al., 2005). Below, we categorize and discuss
different computational methods according to their usage in practice. We will not
specifically differentiate them by the mathematical forms such as univariate or multivariate
models. It should be noted that most of the methods are multivariate and are often used in
combination in practical application.
108 Metabolomics

Biomarker discovery: Like other “omics” studies, the primary objective of many metabolomics
studies is to find biomarkers that are discriminative between matched “case” and “control”
samples, i.e., which metabolites are apparently altered under different physiological
conditions. In pharmaceutical research, metabolomics study has been used for biomarker
discovery for different diseases, safety markers, or drug mechanism research. However,
given the large number of metabolites studied simultaneously with usually small sample
size, it is very common to find metabolites that appear persuasive but in fact spurious. Thus,
it is of crucial importance to control the rate of false positive (Broadhurst & Kell, 2006). To
tackle this problem, many statistical methods have been developed under the term of large
scale hypothesis testing (Benjamini & Hochberg, 1995; Storey 2002; Efron 2003, 2004a, 2004b,
2007a, 2007b, 2008; Storey & Tibshirani, 2003; Reiner et al., 2003; Xie et al., 2005). In classical
hypothesis testing, the fundamental problem is to control type I error, the probability that a
non-trivial finding is declared while it actually happens by chance. Type I error increases
with the number of independent hypothesis considered simultaneously. The well-known
and widely used strategy to control the overall type I error rate is Bonferroni-correction, in
which the critical value for individual hypothesis testing is obtained by dividing the
significance level by the number of hypothesis considered. For example, in metabolomics, if
the search for discriminating biomarkers is performed using 500 metabolites and an
acceptable chance to reject one true hypothesis is 0.05, and then the Bonferroni-corrected
critical value for rejecting an individual null hypothesis for a metabolite should be 0.05/500
= 0.0001). Berferroni-correction is conservative in the sense that it excludes type I errors at
the cost of increasing the potential for type II errors (false negatives) (Broadhurst & Kell,
2006).
A widely accepted error measure in microarray literature for large-scale hypothesis testing
is the false discovery rate (FDR), the proportion of false positives among all the discoveries.
The procedure controlling false discovery rate proposed by Benjamini and Hochberg (1995)
has been recognized as a breakthrough and widely applied or adapted by statistical
researchers (Efron, 2004a). Most of the literatures assume that the theoretical null hypothesis
is known in advance. However, Efron argued that in large scale hypothesis testing, like in
‘omics’ studies, the theoretical null often fails for reasons like correlations among proteins or
genes, unknown confounding factors, or systematic bias (Efron, 2004b, 2007a, 2008). Thus, it
will be appropriate to estimate the distribution of the null statistics from the data in order to
have a more meaningful discovery. Translating to the setting of a metabolomics study,
Efron’s concept aims to find a subset of metabolites that behave very differently from the
majority of the metabolites. Efron’s creative idea has received significant attention from the
research field. Applications to metabolite biomarker finding has not been reported but
certainly expected.
Metabolomics-based biomarker discovery have been reported. In one example for invasive
ovarian carcinomas and ovarian borderline tumors, a differential analysis of 291 detected
metabolites in sixty-six invasive ovarian carcinomas and nine borderline tumors of the
ovary revealed 51 metabolites that were significantly different between borderline tumors
and carcinoma with a FDR controlled at 7.8% (Denkert et al., 2006). For Onchocera volvulus,
analysis of an African sample set of 73 serum and plasm samples based on LC-MS revealed
a set of 14 biomarkers that showed excellent discrimination between Onchocera volvulus-
positive and negative individuals (Denery et al., 2010). Controlling FDR at 54% using
Computational Methods to Interpret and Integrate Metabolomic Data 109

Storey’s q-value approach (Storey, 2002) resulted in 194 features selected from a total of 2350
mass features. Among the 194 features, the authors selected the top 14 feature for
investigation.
Data clustering and visualization: Clustering or unsupervised modeling is useful for class
discovery and provides information on data similarity: metabolomic samples clustered or
grouped together can be objectively considered to be similar.
Principal components analysis (PCA) is probably the most widely used unsupervised
approach to data mining or visualization. PCA is a multivariate technique that transforms
the data into a coordinate system where each new projection (also called principal
components (PC)) is a linear combination of the original variables. PCs are orthogonal so
that each dimension is related to different data characteristics and source of variability in a
mathematical sense (Enot et al., 2008). As a dimension reduction tool, PCA is very useful for
metabolomics data visualization and further data clustering. However, PCA may not work
well for metabolomics data where the differences between groups are minor and obscured
by other covariates.
Self-organizing map (SOM) is another visualization tool for high-dimensional data
(Kohonen, 1998, 2001). The SOM describes a mapping from a higher dimensional input
space to a lower dimensional map space. The procedure for placing a vector from data space
onto the map is to find the node with the closest weight vector to the vector taken from data
space. Once the closest node is located, it is assigned the values from the vector taken from
the data space. The SOM places similar input data in adjacent nodes. Therefore, SOM forms
a semantic map where similar samples are mapped close together and dissimilar apart. One
disadvantage of SOM is that the final map solution is dependent on the order of the
presentation of the training data. The batching-learning version of the algorithm (Kohonen,
2001) overcame this problem and yields reproducible maps. SOM has been applied to
metabolic profiling for clustering blood plasma (Kaartinen et al., 1998), and NMR spectra of
breast cancer tissues (Beckonet et al., 2003b). More recently, Kouskoumvekaki et al. (2008)
applied SOM to identify similarities among the metabolic profiles of different filamentous
fungi. Meinicke et al. (2008) proposed one-dimensional SOM for metabolite-based
clustering and visualization of marker candidates. In a case study on the wound response of
Arabidopsis thaliana, they showed how the clustering and visualization capabilities of SOM
can be utilized to identify relevant groups of biomarkers.
As a popular unsupervised learning method, Hierarchical cluster analysis (HCA) clusters
the data to form a tree diagram or dendrogram which shows the relationships between
samples (Ebbels, 2007). The algorithm begins by computing the distances between all pairs
of samples. Initially each cluster consists of a single sample. The algorithm proceeds
iteratively until all samples are members of a single cluster. The final structure of the
resulting clusters depends on the choice of distance function or “linkage” between two
clusters as well as a similarity cut-off. The most popular choices of linkage are centroidal,
average, single (nearest neighbor) and complete (farthest neighbor) linkages. The centroidal
linkage defines the inter-cluster distance as the distance between the centroids. To
determine the cluster membership, one must decide on a similarity cut-off which breaks the
dendrogram into a number of separate clusters. As an example, Beckobert et al. (2003) used
the HCA method to explore a set of toxicology studies. HCA allowed interpretation of the
110 Metabolomics

data in terms of the magnitude and site of toxicological effect, and helped to explain
misclassifications by other methods.
The k-means clustering is a method of cluster analysis that aims to partition n observations
into k clusters in which each observation belongs to the cluster with the nearest mean. In k-
means clustering, the Euclidean distance is used as a distance metric and variance is used as
a measure of cluster scatter. The number of clusters k is an input parameter. When
performing k-means, it is important to run diagnostic checks for determining the number of
clusters in the data set. Thus, k-means is often used in conjunction with other clustering and
visualization methods (Ebbels, 2007).
Fuzzy k-means (also called Fuzzy c-means) is an extension of k-means clustering technique
based on Fuzzy logic. While k-means discovers hard clusters (a point belong to only one
cluster), Fuzzy k-means is a more statistically formalized method and discovers soft clusters
where a particular point can belong to more than one cluster with certain probability. In
Fuzzy k-means algorithm, one sample can be assigned to more than one class instead of only
one. The membership of each sample is calculated and then represented by a membership
value between 0 and 1, instead of just 0 and 1 in the hard clustering. Cuperlovic-Culf et al.
(2009) presented the application of fuzzy k-means clustering method for the classification of
samples based on metabolomics 1D 1H-NMR fingerprints. The sample classification was
performed on NMR spectra of cancer cell line extracts and of urine samples of type 2
diabetes patients and animal models. The fuzzy k-means clustering method allowed more
accurate sample classification in both datasets relative to the other tested methods including
PCA, HCA and k-means clustering. Li et al. (2009) applied fuzzy k-means to cluster three
gene types of Escherichia coli on the basis of their metabolic profiles and delivered better
results than PCA. On the basis of the optimized parameters, the fuzzy k-means was able to
reveal main phenotype changes and individual characters of three gene types of E. coli,
while PCA failed to model the metabolite data.
Clustering of metabolomics data can be hampered by noise originating from biological
variation, physical sampling error and analytical error. Bootstrap aggregating (bagging) is a
resampling technique that can deal with noise and improves accuracy. Hageman et al.
(2006) demonstrated the application of bagged clustering to metabolomics data. It was
argued that the bagged k-means should be favored against ordinary k-means clustering
when dealing with noisy metabolomics data.
In practice, it is common to combine dimensionality reduction and clustering methods. For
example, first, a sample-based principal component analysis (PCA) is performed to compute
a subset of principal components. Then the metabolite-specific PCA loadings of these
components are used for metabolite-based clustering using k-means or hierarchical methods
(Pohjanen et al., 2006).
Classification and prediction: While the purpose of clustering is to group similar data together,
classification aims at finding a rule to discriminate the classes in an optimal way as well as
selecting the subset of features that are most discriminative or predicative. In contrast to
clustering applications, the class label and the number of classes are known for a subset of
data (training samples) in a priori in classification problem. Once the rule or classifies are
determined using training dataset, it can be used to predict the class label (such as diseased
or not) of a test sample.
Computational Methods to Interpret and Integrate Metabolomic Data 111

The k-Nearest Neighbour (kNN) rule for classification may be the simplest of all supervised
classification approaches (Ebbels, 2007). Different from other supervised learning methods,
the training phase of kNN consists of only storing the training samples and the
corresponding class labels. In the classification (prediction) phase, a test sample is classified
by assigning the label which is most frequent among the k nearest training samples. The
method requires only the choice of k, the number of neighbors to be considered when
making the classification. Greater values of k reduce the effect of noise on the classification,
but make boundaries between classes less distinct. A limitation of this classification method
is that the classes with the more frequent examples tend to dominate the prediction of a test
sample. Usually, k is chosen through a cross validation procedure. kNN has been often used
as a comparator for other methods in literature (Beckonert et al., 2003; Baumgartner et al.,
2004).
Partial least square for discriminant analysis (PLS-DA) is a regression extension of PCA that
takes advantage of class label information to maximize separation between groups of
observations. PLS-DA models the relationship between the class affiliation matrix (Y) and
feature matrix (X), which is a generalization of multiple linear regressions. It determines a
set of latent variables explaining as much as possible of the covariance between the two
matrices. PLS-DA can deal with uncompleted dataset and multicollinearity problem. The
output of PLS-DA is the score matrix that can be plotted similarly as in PCA and the
predictor matrix containing estimated class affiliation (Ciosek et al., 2005; Trygg &
Lundstedt, 2007). The ortogonal-PLS (OPLS) method (Cloarec et al., 2005; Trygg &
Lundstedt, 2007) is a recent modification of the PLS method. The main idea of OPLS is to
separate the systematic variation in X into two parts, one that is linearly related to Y and one
that is orthogonal to Y. The OPLS method provides a prediction similar to that of PLS.
However, the interpretation of the models is improved because the structured noise is
modeled separately from the variation common in X and Y. Analogous to PLS, when the Y
matrix is class affiliation, the corresponding analysis is named OPLS-DA. Cloarec et al.
(2005) illustrated the applicability of the method in combination with statistical total
correlation spectroscopy to 1H NMR spectra of urine from a metabonomic study of a model
of insulin resistance based on the administration of a carbohydrate diet to three different
mice strains. Tapp and Kemsley (2009) recently discussed similarities and differences
between PLS-DA and OPLS-DA with a focus on the usage of OPLS in the analytical
chemistry literature. They concluded that the two methods are very similar and no one
outperforms the other, and the reported discrepancies in the literature must be due to
differences in the implementation details, or some otherwise ‘‘unfair’’ comparison between
the methods
An Artificial neural network (ANN) is a widely used non-linear data modeling tool (Bishop,
1995). An ANN is a computational model that is inspired by the structure and functional
aspects of biological neural network. An ANN consists of a layered network of nodes with
simple linear or sigmoid activation function. The most widely used type of ANN is the
multilayer perceptron (MLP), which has at least three layers including the input layer, one
or more hidden layers and output layer. The most attractive feature of a MLP is its capacity
to approximate any continuous function in arbitrary precision given enough number of
nodes in the hidden layers with sigmoid-type activation functions. ANN has been applied to
classification of tumor cells by different researchers (Maxwell et al., 1998; Ott et al., 2003).
112 Metabolomics

One disadvantage of ANN is that non-linear optimization algorithm is needed to train a


neural network. It is easy for the optimization procedure to be stuck in local minima while
finding the optimal parameters. Thus, global optimal solution is not guaranteed. Moreover,
it is difficult to interpret the connection weights to gain some biological insight for feature
ranking in applications.
Support vector machines (SVMs) is a supervised learning technique for classification and
regression (Cristianini & Shawe-Taylor, 2000). A support vector machine constructs an
optimal hyperplane for classification. The hyperplane is constructed in such a way that it
has the largest distance to the nearest training data points of any class. In contrast to
ANN, SVMs is trained by using convex programming such as quadratic programming.
Thus, it can find the global optimal solution efficiently. The computation complexity of
SVMs depends on the number of samples instead of the dimension of each sample. Thus
there is no curse of dimensionality problem. When cases are not linearly separable,
appropriate kernel functions can be chosen to transform the original data into high
dimensional feature space. SVMs is well known for its good generalization capacity. For
nearly a decade, SVMs has been used in the field of bioinformatics for classifying and
evaluating gene expression microarray data (Furey et al., 2000; Guyon et al., 2002).
Mahadevan et al. (2008) compared the performance of PLS-DA multivariate analysis to
SVMs and showed that SVMs were superior to PLS-DA in terms of predictive accuracy
with the least number of features. With fewer features, SVMs were able to give better
predictive model when compared to that of PLS-DA.
Computation methods for identification of metabolites: Metabolomics studies are targeted at
identifying and quantifying all metabolites in a given biological context. One central
problem is the translation of the measured mass into molecular formulae. Given the
observed mass and atoms, the first problem is to find all the possible elemental
compositions by solving the Diophantine equation:

n m
i
i i M ,

where M is the observed mass and mi is the mass of the ith atom. Diophantine equation is
the basis for much of the mass spectrometry software to obtain compositions. Usually, there
are many integer solutions to it mathematically. Among all the mathematical solutions, we
then seek all of the integers ni that are chemically feasible considering some chemical
contextual information such as the valence rules, double-bond equivalents or exact mass
(Meija, 2006). Even with very high mass accuracy (<1 ppm), many chemically possible
formulae can be obtained in higher mass regions. To further reduce the number of potential
elemental compositions, it is necessary to utilize isotope abundance pattern (Kind & Fiehn,
2006).
The identification of small metabolites has been seen as one of the bottlenecks in
interpreting metabolomics data. Neumann & Bocker (2010) provided a review focusing on
the computational methods for electronspary ionization (ESI) mass spectrometry. One of the
most common methods for the identification of compounds using mass spectrometry is the
comparison with spectra of authentic standards. The Metabolomics Standards Initiative
Computational Methods to Interpret and Integrate Metabolomic Data 113

(MSI) has defined several confidence levels for the identification of non-novel chemical
compounds, ranging from level 1 for a rigorous identification to unidentified signals at level
4 (Sumner et al., 2007). The difference between level 1 and 2 is that the former requires the
comparison with authentic standards based on in-house data measured under identical
analytical conditions, whereas the latter allows one to use literature or external databases.
Level 1 or level 2 identifications are based on a comparison of “exact mass and isotope
pattern”. Even with the most exact mass and isotope pattern the identification will be
limited to the elemental composition. Many compounds share the same sum formula for
known metabolites in databases such as KEGG or PubChem. For all other MSI levels, the
“identification” usually reduces to an annotation with lower levels of confidence (Neumann
& Bocker, 2010).
When a reference spectrum is used, a similarity or distance function is needed for selecting
database entries. The most basic similarity functions are those based on counting the
number of matching peaks between a query spectrum and each of the database spectra. For
this, both spectra can be considered as binary vectors with 0’s and 1’s for “peak absent” and
“peak present”, respectively (Neumann & Bocker, 2010). Common distance functions on
binary vectors are the Hamming distance (counting any difference) or the Jaccard coefficient
(the fraction of matching peaks). Besides counting matches, other measures also consider
their actual mass and intensity, such as the Euclidean distance, the probability-based
matching (PBM), the normalized dot product (NDP), and a modified cosine distance for the
database search of EI spectra (Stein, 1994).
Oberacher et al. (2009) proposed and optimized a search function for tandem mass
spectrometry (MS/MS-spectra) based on a combination of relative and absolute match
probabilities, which combines the principle of peak counting and summed intensities of
matching peaks. The X-Rank algorithm (Mylonas et al., 2009) for MS/MS-spectra match is
based on probability calculations. It sorts peak intensities of a spectrum and then establishes
a correlation between two sorted spectra. X-Rank computes the probability that a rank from
an experimental spectrum matches a rank from a reference library spectrum. The solution
requires training on a representative dataset. In a training step, characteristic parameter
values are generated for a given data set. Identification of small compounds is still
challenging, especially for compounds that have not been recorded in any library or
structure database. Methods for these tasks are highly sought.

3.2 Computational methods for metabolic pathway analysis using metabolomic data
Metabolomics data provides a series of snapshots of cellular metabolism, which can be
combined with metabolic flux data for further analysis. Metabolic pathways are the true
functional units of metabolic systems (Schilling et al., 2000b). Finding biochemically
plausible pathways between metabolites in a metabolic network is a central problem in
computational metabolic modeling. Mathematical modeling approaches to metabolic
regulation analysis involve different levels of details and complexities ranging from detailed
kinetic models, stoichiometric analysis, structural kinetic models, to large scale topological
network analysis (Steuer, 2007).
Detailed kinetic models of metabolic pathways, based on explicit enzymekinetic rate
equations, is a bottom-up approach towards more comprehensive large-scale dynamic
114 Metabolomics

models. It allows for the most detailed quantitative evaluation of the dynamics of metabolic
systems, which is very important for improving the understanding of metabolic regulation
and control. The metabolic control analysis (MCA) is the culminating mathematical theory
from kinetic models, which describes the control and regulatory properties of metabolic
systems (Heinrich & Schuster, 1996; Fell, 1997).
A metabolic network is a collection of enzyme-catalyzed reactions and transport processes
that serve to dissipate substrate metabolites and generate final metabolites. The dynamics of
a metabolic system can be described by a set of ordinary differential equations:

dXi
  Sij v j ( X , k )  S  v( X , k ), (1)
dt j

where X i represents the concentration of the metabolite and Sij stands for the
stoichiometric coefficient for the reactant i in the jth reaction. v j ( X , k ) corresponds to the
flux through the jth reaction. The vector v( X , k ) consists of nonlinear enzyme-kinetic rate
functions, which depends on the concentration X and a set of kinetic parameters k . Given
the v( X , k ) function form, the set of kinetic parameters k and an initial state X (0) , the
differential equations can be solved numerically to obtain the time-dependent behavior of
all metabolites under consideration. The stoichiometric matrix S is an m by n matrix where
m corresponds to the number of metabolites and n is the total number of metabolites and n
is the total number of fluxes taking place in the network.
The stoichiometric analysis approach takes advantage of the structure nature of metabolic
system. Knowledge of the stoichiometry puts constraints on the feasible flux distributions,
which can be utilized to model the functional capabilities of metabolic networks (Varma &
Palsson 1994; Edwards & Palsson, 2000; Stelling et al., 2002; Famili et al., 2003; Price et al.,
2003).
The pathway structure should be an invariant property of the network along with
stoichiometry. Under steady-state, the set of ordinary equations reduce to linear
equations:

0  S  v( X 0 , k )  S  v 0 , (2)

which is typically underdetermined as the number of reactions exceeds the number of


participating metabolites. The set of all solutions v 0 to the steady state equation (2) is
called null space. A set of basis vectors corresponding to a steady-state biochemical
pathway can be selected to describe the null space. Additional constraints from
biological insights are necessary to determine the system completely (Schilling et al.,
2000b), which generally results in a set of linear equalities and inequalities. The set of
feasible solutions under bio-chemical constraints form a convex steady-state flux cone.
The convex analysis of biochemical networks was founded by Clarke (Clarke, 1980,
1988). The study of convex flux cones utilizes methods and concepts rooting in linear
algebra and optimization and forms the underlying mathematical structure for metabolic
pathway analysis.
Computational Methods to Interpret and Integrate Metabolomic Data 115

Flux balance analysis (FBA) is a computational approach to reduction of the admissible flux
space (Schilling et al., 1999; Edwards & Palsson, 2000). FBA optimizes an objective function
such as maximal biomass yield or maximal energy production through the steady-state flux
space has resulted in many applications (Papin et al., 2004; Almaas et al., 2004;
Stephanopoulos et al., 2004). Using FBA, in silico studies on the systemic properties of the
Haemophilus influenzae and E. coli (Edwards & Palsson, 2000) metabolic networks have been
completed. Under various substrate conditions, Schilling et al. (2000a) explored the
metabolic capabilities and predicted functions of a sub-system of the E. coli using FBA.
A metabolic network can be decomposed into distinct pathways, termed elementary flux
modes (EFM). An EFM is the minimal set of reactions capable of working together in a
steady state, which is unique for a given metabolic network. Another closely related concept
is extreme pathways, which are a subset of elementary modes (Klamt & Stelling, 2003). All
feasible flux vectors can be described as linear combinations of EFMs. The concept of EFM
has resulted in a vast number of applications for metabolic network analysis (Stelling et al.,
2002; Schuster et al., 2002; Klamt & Schuster, 2002; Klamt & Gilles, 2004; Klamt et al., 2006).
For medium-sized metabolic networks, software packages have been developed for the
computation of elementary flux modes (Hoops et al., 2006; Klamt et al., 2007). Owing to a
combinatorial explosion of the number of elementary vectors, this approach becomes
computationally intractable for genome scale networks. To develop an analysis approach
computationally feasible even for genome scale networks, Urbanczik & Wagner (2005)
proposed to focus on conversion cone, the projection of the flux cone, which describes the
interaction of the metabolism with its external chemical environment. The method for
calculating the elementary vectors of this cone was applied to study the metabolism of
Saccharomyces cerevisiae.
Stoichiometric analysis does not incorporate dynamic properties into the description of the
system. Steuer (2006, 2007) proposed a structure kinetic modeling approach to augmenting
the stoichiometric analysis with kinetic properties. The idea of the proposed approach is to
use a local linear approximation to explicit kinetic model to capture the dynamic response to
perturbations, the stability of a metabolic state, as well as the transition to oscillatory
behavior. The local linear approximation is obtained from a Taylor series expansion of the
metabolic system. The linear term of the expansion is the derivative of the kinetic rate
equations with respect to the metabolic concentration X at a given state, which usually
requires knowledge of the enzyme-kinetic rate equations. Even in the absence of enzyme-
kinetic information, it is still possible to specify the structure of the linear term. Structure
kinetic modeling approach allows quantitative conclusions about the possible dynamics of
the system, based on only a minimal amount of additional information.
The extension of the detailed kinetic models to whole cell models is faced with some
fundamental difficulties including the absence of comprehensive measured kinetic
parameter values, and the observed inconsistency in the available kinetic data, and the
computational complexity of such models. Traditionally, kinetic models are constructed
using rate equations derived to describe conditions in vitro and thus rely on the use of in
vitro measured kinetic parameters. However, the conditions at which in vitro experiments
are performed are often very different from those inside the cell. Thus, in vitro kinetic rates
and in vitro kinetic parameters describe enzymatic behaviors that may not truly represent
the observed physiological kinetic behavior in the cell. Several methods have been proposed
116 Metabolomics

to address this issue by incorporating in vivo measurements in constructing kinetic models


(Visser & Heijnen, 2003) or estimating kinetic parameters in biochemical networks using
measured variables (Lei & Jorgensen, 2001; Moles et al., 2003; Segre et al., 2003). These
methods require considerable mathematical efforts or utilize nonlinear optimization
techniques. (Famili et al., 2003) proposed an approach for incorporating steady-state in vivo
data with constraint based modeling approach to determine all candidate numerical values
of kinetic constants. The kinetic solution space, termed k-cone, contains all the allowable
numerical values of the kinetic constants. The k-cone is obtained by approximating the
nonlinear kinetic rate laws as a linear or bilinear function of the kinetic constants. The k-cone
approach can be used to determine consistency between in vitro measured kinetic values
and in vivo concentration and flux measurements when used in a network-scale kinetic
model. To calculate the relationship between kinetic parameters measured in vitro and the
k-cone, optimization methods were essential. It was successful in determining whether in
vitro measured kinetic values used in the reconstruction of a kinetic-based model of
Saccharomyces cerevisiae central metabolism could reproduce in vivo measurements.
Despite the number of research published, information gained from theoretical or
experimental metabolic network has not fully enabled probing biochemical pathway
structure with the aim at detecting novel metabolic routes (Fiehn, 2007).

4. Computational methods to integrate metabolomic data with other “omics”


datasets
It is also becoming clear that any single “omics” approach may not be sufficient to
characterize the complexity of biological systems and an integrated “omics” approach may
be a key to decipher complex biological systems (Gygi et al., 1999; Zhang et al., 2010). In
general, integrated analysis of metabolomics dataset with other types of “omics” datasets
can increase both dimension of information sources and statistical power in order to
generate a conclusion with high confidence. In recent years, several computational methods
have been applied to integrate metabolomics with other “omics” datasets, and the results
demonstrated that better pattern recognition and association identification can be achieved
when proper mathematical, statistical and other computational tools were applied.
The simple correlation analysis, Pearson or Spearman’s correlation has been used to assess
degree of association between metabolomic and transcriptomic data. For example, it has
been used to distinguish different potato tuber systems, and to determine the relationship
between genes and their paired metabolites (Urbanczyk-Wochniak et al., 2003, 2007),
through integrating metabolomic and transcriptomic datasets.
Because of the high dimensionality involved in the metabolomic data and other “omics”
data, the dimension reduction tool such as PCA can demonstrate its power in the integrated
analysis. It has been used in two different but related scenarios: i) PCA applies to each
“omics” data to reduce the dimensionality of each omics data so that the “omics” data are
ready for integration; ii) PCA applies to the integrated metabolomics and other “omics” data
directly to identify a particular pattern. In scenario 1, metabolomic data or other “omics”
data are not directly used in any integrated analysis. Rather, PCA was used to reduce the
dimension of each data, so that researchers can focus on the most important components of
metabolomic data or other “omics” data (Urbanczyk-Wochniak et al., 2003; Rubingh et al.,
Computational Methods to Interpret and Integrate Metabolomic Data 117

2009; van den Berg et al., 2009). In scenario 2, PCA was directly used to identify pattern of
the integrated metabolomic data with other “omics” data. For example, it was used to a
integrated metabolomic data and a proteomic data to reveal clustering of the two genotypes
(Weckwerth et al., 2004). While PCA can also be used for exploring polynomial relationships
and for multivariate outlier detection, this method is restricted to linear relationships.
In both correlation and PCA analysis, the roles of all variables are the same and they are
interchangeable. They are used to explore associations between factors. In some other
analyses, some factors (independent variables X) are used to explain or predict the variable
of main interest (dependent variable Y). For example, PLS is a statistical method that models
Y over X through a linear relationship. Rather than considering all dependent variables as
regressors in a multivariate regression analysis, PLS regresses Y over principal components
resulted from a principal components analysis (Garthwaite 1994). The method was in fact
previously used to model metabolomic variables as a function of the transcriptome profiles
(Pir et al., 2006). The analysis allowed the discrimination between the effects that the growth
media, dilution rate and deletion of specific genes on the transcriptomic and metabolomic
profiles (Pir et al. 2006). The method was also used to relate quantifiable phenotypes of
interest such as protease activity or productivity, to concentrations of each of the metabolites
determined (Braaksma et al., 2011). The analysis revealed various sugar derivatives
correlated with glucoamylase activity.
As an extension of PLS, Le Cao and colleagues proposed a sparse PLS approach to combine
integration and simultaneous variable (e.g., gene) selection in one step (Le Cao et al., 2008,
2009). In the approach, the PLS was penalized by the sum of the absolute values of the
coefficients through least absolute shrinkage and selection operator (LASSO) (Tibshirani
1996), therefore automatically eliminating variables (e.g., genes) with negligible effects. The
model selection approach, together with the smoothly clipped absolute deviation approach
(Fan & Li, 2001) is effective in analyzing data with sparsity (e.g., only a few genes have
significant effects).
The methods previously discussed in this section, including Perason correlation, PCA, and
PLS, are all methods to explore linear relationship. On the other hand, the kinetic model and
artificial network could be more sensible when nonlinearity occurs. In an analysis to
integrate metabolomics and pharmacokinetics (or nutrikinetics), Van Velzen et al. (2009)
presented a one-compartment nutrikinetic model with first-order excretion, a lag time, and a
baseline function was fitted to the time courses of these selected biomarkers based on
metabolomic data. A kietic model was also used to model the relationship between enzyme
kinetics and intracellular metabolites through a two-substrate Michaelis–Menten equation
with competitive substrate inhibition or competitive product inhibition (Schroer et al., 2009).
The kinetic constants were estimated by nonlinear regression of initial rate measurements.
Martense & Vanrolleghem (2010) summarized a few other modeling approaches. Compared
to data driving unsupervised analysis, the mathematical modeling may provide a meaning
relationship for a better understanding. However, because the modeling is generally based
on approximation under some restricted assumptions, the simple model may not precisely
describe the complex biology system.
Artificial network is another method to cope with the nonlinearity. A batch-learning self-
organizing network was utilized to classify the metabolomes and the transcriptomes
118 Metabolomics

according to their time-dependent pattern of changes (Kanaya et al., 2001); the results
showed that the metabolomes and transcriptomes regulated by the same mechanism tended
to be clustered together (Hirai et al., 2004, 2005). The A batch-learning self-organizing
network is artificial neural network that is trained using unsupervised learning to produce a
low-dimensional representation of the input space of the training samples. A Network-
embedded thermodynamic analysis (NET analysis) is presented as a framework for
mechanistic and model-based analysis of metabolite data. By coupling the data to an
operating metabolic network via the second law of thermodynamics and the metabolites’
Gibbs energies of formation, NET analysis allows inferring functional principles from
quantitative metabolite data; for example it identifies reactions that are subject to active
allosteric or genetic regulation as exemplified with quantitative metabolomic data from E.
coli and S. cerevisiae (Kummel et al., 2006). The network typically creates a graphic
representing the global relationship. In a review article, Feist et al. (2006) classified studies
using network into three categories: studies that use a reconstruction to examine topological
network properties, studies that use a reconstruction in constraint-based modeling for
quantitative or qualitative analyses, and studies that are purely data driven. Some of the
networks’ mathematical frameworks are graph theory. It provides a visual presentation of
the complex biology system. However, when it involves more features, the network
approach often become too complicated to provide a clear clue.
Bayesian graphical modeling approaches infer biological regulatory networks by integrating
expression levels of different types. Specific sequence/structure information will be
incorporated into the prior probability models (Webb-Robertson et al., 2009) presented a
Bayesian approach to integration that uses posterior probabilities to assign class memberships
to samples using individual and multiple data sources; these probabilities are based on lower-
level likelihood functions derived from standard statistical learning algorithms. The approach
was demonstrated by integrating two proteomic datasets and one metabolic dataset from
microbial infections of mice; the results showed that integration of the different datasets
improved classification accuracy to ~89% from the best individual dataset at ~83 %.
Integrative interpretations of data from different “omics” including metabolomics, are still
in it early development stage. More thoughtful interpretation methods that are capable to
reveal biology at a system level are yet to come. The collaboration between mathematicians,
statisticians, bioinformaticians and experimental biologists will be the key to the success of
these efforts.

5. Final remarks
Although comprehensive coverage of metabolome in cells is not yet possible, significant
advancements in the large-scale profiling of metabolites have been achieved in recent years
and these analyses have offered unique insight into the metabolic and regulatory networks
of cells. In this chapter, we first reviewed some of the widely used and emerging
technologies for metabolomics analysis, and then focus on recent progress in developing
computational methodologies to improve biological interpretation of high throughput
metabolomic data. In addition, we present some mathematical, statistical and bioinformatics
methods that have been utilized for the integration of metabolomics data with other type of
“omics” datasets and how this integrative analysis has improved our interpretation of
biological systems.
Computational Methods to Interpret and Integrate Metabolomic Data 119

6. Acknowledgements
The research of J. Wang and W. Zhang of Tianjin University was supported by National
Basic Research Program of China (National “973” program, project No. 2011CBA00803 and
No. 2012CB721101), and National Natural Science Foundation of China (Project No.
31170043).

7. Disclaimer
Feng Li is currently working for Office of Biostatistics, Food and Drug Administration.
Views expressed in this paper are the author's professional opinions and do not necessarily
represent the official positions of the U.S. Food and Drug Administration

8. References
Aharoni, A., C. H. R. De Vos, H. A. Verhoeven, C. A. Maliepaard, G. Kruppa, R. Bino & D. B.
Goodenowe (2002) Nontargeted metabolome analysis by use of fourier transform
ion cyclotron mass spectrometry. OMICS A Journal of Integrative Biology, 6, 217-234.
Almaas, E., B. Kovacs, T. Vicsek, Z. N. Oltvai & A. L. Barabasi (2004) Global organization of
metabolic fluxes in the bacterium Escherichia coli. Nature, 427, 839-843.
Amantonico, A., P. L. Urban, J. Y. Oh & R. Zenobi (2009) Interfacing microfluidics and laser
desorption/Ionization mass spectrometry by continuous deposition for application
in single cell analysis. Chimia, 63, 185-188.
Amantonico, A., J. Y. Oh, J. Sobek, M. Heinemann & R. Zenobi (2008) Mass spectrometric
method for analyzing metabolites in yeast with single cell sensitivity. Angewandte
Chemie-International Edition, 47, 5382-5385.
Amantonico, A., P. L. Urban & R. Zenobi (2010) Analytical techniques for single-cell
metabolomics: state of the art and trends. Analytical and Bioanalytical Chemistry, 398,
2493-2504.
Anderle, M., S. Roy, H. Lin, C. Becker & K. Joho (2004) Quantifying reproducibility for
differential proteomics: noise analysis for protein liquid chromatography-mass
spectrometry of human serum. Bioinformatics, 20, 3575-3582.
Baumgartner, C., C. Bohm, D. Baumgartner, G. Marini, K. Weinberger, B. Olgemoller, B.
Liebl & A. A. Roscher. 2004. Supervised machine learning techniques for the
classification of metabolic disorders in newborns. In Bioinformatics, 2985-96.
England.
Beckonert, O., M. E. Bollard, T. M. D. Ebbels, H. C. Keun, H. Antti, E. Holmes, J. C. Lindon &
J. K. Nicholson (2003) NMR-based metabonomic toxicity classification: hierarchical
cluster analysis and k-nearest-neighbour approaches. Analytica Chimica Acta, 490, 3-
15.
Beckonert, O., H. C. Keun, T. M. D. Ebbels, J. G. Bundy, E. Holmes, J. C. Lindon & J. K.
Nicholson (2007) Metabolic profiling, metabolomic and metabonomic procedures
for NMR spectroscopy of urine, plasma, serum and tissue extracts. Nature Protocols,
2, 2692-2703.
Bedair, M. & L. W. Sumner (2008) Current and emerging mass-spectrometry technologies
for metabolomics. Trac-Trends in Analytical Chemistry, 27, 238-250.
120 Metabolomics

Benjamini, Y. & Y. Hochberg (1995) Controlling the False Discovery Rate: A Practical and
Powerful Approach to Multiple Testing. Journal of the Royal Statistical Society. Series
B (Methodological), 57, 289-300.
Berg, J., Y. P. Hung & G. Yellen (2009) A genetically encoded fluorescent reporter of
ATP:ADP ratio. Nature Methods, 6, 161-166.
Birkemeyer, C., A. Kolasa & J. Kopka (2003) Comprehensive chemical derivatization for gas
chromatography-mass spectrometry-based multi-targeted profiling of the major
phytohormones. Journal of Chromatography A, 993, 89-102.
Bishop, C. M. 1995. Neural Networks for Pattern Recognition. Oxford, UK: Clarendon Press.
Braaksma, M., S. Bijlsma, L. Coulier, P. J. Punt & M. J. van der Werf (2011) Metabolomics as
a tool for target identification in strain improvement: the influence of phenotype
definition. Microbiology, 157, 147-59.
Breitling, R., A. R. Pitt & M. P. Barrett (2006) Precision mapping of the metabolome. Trends
in Biotechnology, 24, 543-548.
Broadhurst, D. & D. Kell (2006) Statistical strategies for avoiding false discoveries in
metabolomics and related experiments. Metabolomics, 2, 171-196.
Buckmaster, R., F. Asphahani, M. Thein, J. Xu & M. Zhang (2009) Detection of drug-induced
cellular changes using confocal Raman spectroscopy on patterned single-cell
biosensors. Analyst, 134, 1440-1446.
Cahill, G., P. K. Walsh & D. Donnelly (2000) Determination of yeast glycogen content by
individual cell spectroscopy using image analysis. Biotechnology and Bioengineering,
69, 312-322.
Cannon, D. M., N. Winograd & A. G. Ewing (2000) Quantitative chemical analysis of single
cells. Annual Review of Biophysics and Biomolecular Structure, 29, 239-263.
Chen, H. W., A. Venter & R. G. Cooks (2006) Extractive electrospray ionization for direct
analysis of undiluted urine, milk and other complex mixtures without sample
preparation. Chemical Communications, 2042-2044.
Ciosek, P., Z. Brzozka, W. Wroblewski, E. Martinelli, C. Di Natale & A. D'Amico. 2005.
Direct and two-stage data analysis procedures based on PCA, PLS-DA and ANN
for ISE-based electronic tongue-Effect of supervised feature extraction. In Talanta,
590-6. England.
Clarke, B. L. 1980. Stability of Complex Reaction Networks. In Advances in Chemical Physics,
1-215. John Wiley & Sons, Inc.
--- (1988) Stoichiometric network analysis. Cell Biophysics, 12, 237-53.
Cloarec, O., M. E. Dumas, A. Craig, R. H. Barton, J. Trygg, J. Hudson, C. Blancher, D.
Gauguier, J. C. Lindon, E. Holmes & J. Nicholson (2005) Statistical total correlation
spectroscopy: an exploratory approach for latent biomarker identification from
metabolic 1H NMR data sets. Analytical Chemistry, 77, 1282-9.
Cooks, R. G., Z. Ouyang, Z. Takats & J. M. Wiseman (2006) Ambient mass spectrometry.
Science, 311, 1566-1570.
Cristianini, N. & J. Shawe-Taylor. 2000. n Introduction to Support Vector Machines. Cambridge
University Press.
Cuperlovic-Culf, M., N. Belacel, A. S. Culf, I. C. Chute, R. J. Ouellette, I. W. Burton, T. K.
Karakach & J. A. Walter (2009) NMR metabolic analysis of samples using fuzzy K-
means clustering. Magnetic Resonance in Chemistry, 47 Suppl 1, S96-104.
Daykin, C. A., P. J. D. Foxall, S. C. Connor, J. C. Lindon & J. K. Nicholson (2002) The
comparison of plasma deproteinization methods for the detection of low-
Computational Methods to Interpret and Integrate Metabolomic Data 121

molecular-weight metabolites by H-1 nuclear magnetic resonance spectroscopy.


Analytical Biochemistry, 304, 220-230.
Denery, J., A. Nunes, M. Hixon, T. Dickerson, K. Janda (2010) Metabololics-based discovery
of diagnostic biomarkers for onchoceriasis. PLoS Neglected Tropical Diseases,
4(10), e384.
Deng, C. H., X. M. Zhang & N. Li (2004) Investigation of volatile biomarkers in lung cancer
blood using solid-phase microextraction and capillary gas chromatography-mass
spectrometry. Journal of Chromatography B-Analytical Technologies in the Biomedical
and Life Sciences, 808, 269-277.
Denkert, C., J. Budczies, T. Kind, W. Weichert, P. Tablack, J. Sehouli, S. Niesporek, D.,
Konsgen, M. Dietel, and O. Fiehn (2006) Mass Spectrometry–Based Metabolic
Profiling Reveals Different Metabolite Patterns in Invasive Ovarian Carcinomas
and Ovarian Borderline TumorsSpec. Cancer Res, 66: (22).
de Oliveira, L. F. C., R. Colombara & H. G. M. Edwards (2002) Fourier transform Raman
spectroscopy of honey. Applied Spectroscopy, 56, 306-311.
Dettmer, K., P. A. Aronov & B. D. Hammock (2007) Mass spectrometry-based metabolomics.
Mass Spectrometry Reviews, 26, 51-78.
De Vos, R. C. H., S. Moco, A. Lommen, J. J. B. Keurentjes, R. J. Bino & R. D. Hall (2007)
Untargeted large-scale plant metabolomics using liquid chromatography coupled
to mass spectrometry. Nature Protocols, 2, 778-791.
Dieterle, F., A. Ross, G. Schlotterbeck & H. Senn (2006) Metabolite projection analysis for fast
identification of metabolites in metabonomics. Application in an amiodarone study.
Analytical Chemistry, 78, 3551-61.
Dunn, W. B., N. J. C. Bailey & H. E. Johnson (2005) Measuring the metabolome: current
analytical technologies. Analyst, 130, 606-625.
Dunn, W. B. & D. I. Ellis (2005) Metabolomics: current analytical platforms and
methodologies. Trac-Trends in Analytical Chemistry, 24, 285-294.
Ebbels, T. M. D. 2007. Non-linear Methods for the Analysis of Metabolic Profiles. In The
Handbook of Metabonomics and Metabolomics, eds. J. C. Lindon, J. K. Nicholson & E.
Holmes, 201--226. London: Elsevier.
Edwards, J. S. & B. O. Palsson. 2000. The Escherichia coli MG1655 in silico metabolic
genotype: its definition, characteristics, and capabilities. In Proceedings of the
National Academy of Sciences of the United States of America, 5528-33. United States.
Efron, B. (2003) Robbins, Empirical Bayes and Microarrays. The Annals of Statistics, 31, 366-
378.
--- (2004a) Large-Scale Simultaneous Hypothesis Testing. Journal of the American Statistical
Association, 99, 96-104.
--- (2004b) Large-Scale Simultaneous Hypothesis Testing: The Choice of a Null Hypothesis.
Journal of the American Statistical Association, 99, 96-104.
--- (2007a) Correlation and Large-Scale Simultaneous Significance Testing. Journal of the
American Statistical Association, 102, 93-103.
--- (2007b) Size, power and false discovery rates. The Annals of Statistics, 35, 1351-1377.
--- (2008) Microarrays, Empirical Bayes and the Two-Groups Model. Statistical Science, 23, 1-
22.
Enot, D. P., B. Haas & K. M. Weinberger (2011) Bioinformatics for mass spectrometry-based
metabolomics. Methods in Molecular Biology, 719, 351-75.
122 Metabolomics

Enot, D. P., W. Lin, M. Beckmann, D. Parker, D. P. Overy & J. Draper (2008) Preprocessing,
classification modeling and feature selection using flow injection electrospray mass
spectrometry metabolite fingerprint data. Nature Protocols, 3, 446-70.
Famili, I., J. Forster, J. Nielsen & B. O. Palsson. 2003. Saccharomyces cerevisiae phenotypes can
be predicted by using constraint-based analysis of a genome-scale reconstructed
metabolic network. In Proceedings of the National Academy of Sciences of the United
States of America, 13134-9. United States.
Fan, J. Q. & R. Z. Li (2001) Variable selection via nonconcave penalized likelihood and its
oracle properties. Journal of the American Statistical Association, 96, 1348-1360.
Farag, M. A., D. V. Huhman, R. A. Dixon & L. W. Sumner (2008) Metabolomics reveals
novel pathways and differential mechanistic and elicitor-specific responses in
phenylpropanoid and isoflavonoid biosynthesis in Medicago truncatula cell
cultures. Plant Physiology, 146, 387-402.
Fehr, M., W. B. Frommer & S. Lalonde (2002) Visualization of maltose uptake in living yeast
cells by fluorescent nanosensors. Proceedings of the National Academy of Sciences of the
United States of America, 99, 9846-9851.
Feist, A. M., J. C. Scholten, B. O. Palsson, F. J. Brockman & T. Ideker (2006) Modeling
methanogenesis with a genome-scale metabolic reconstruction of Methanosarcina
barkeri. Molecular Systems Biology, 2, 2006 0004.
Fell, D. 1997. Understanding the Control of Metabolism. Potland Press.
Fiehn, O. (2001) Combining genomics, metabolome analysis, and biochemical modelling to
understand metabolic networks. Comparative and Functional Genomics, 2, 155-168.
Fiehn, O. 2007. Cellular Metabolomics: The Quest for Pathway Structure. In The Handbook of
Metabonomics and Metabolomics, eds. J. C. Lindon, J. K. Nicholson & E. Holmes, 35-
54. Elsevier.
Fletcher, J. S. (2009) Cellular imaging with secondary ion mass spectrometry. Analyst, 134,
2204-2215.
Fliermans, C. B. & E. L. Schmidt (1975) Authoradiography and immunofluorescence
combined for autecological study of single cell activity with nitrobacter as a model
system. Applied Microbiology, 30, 676-684.
Fraser, P. D., E. M. A. Enfissi, M. Goodfellow, T. Eguchi & P. M. Bramley (2007) Metabolite
profiling of plant carotenoids using the matrix-assisted laser desorption ionization
time-of-flight mass spectrometry. Plant Journal, 49, 552-564.
Furey, T. S., N. Cristianini, N. Duffy, D. W. Bednarski, M. Schummer & D. Haussler (2000).
Support vector machine classification and validation of cancer tissue samples using
microarray expression data. Bioinformatics, 16, 906-914.
Garthwaite, P. H. (1994) An Interpretation of Partial Least-Squares. Journal of the American
Statistical Association, 89, 122-127.
Gmati, D., J. K. Chen & M. Jolicoeur (2005) Development of a small-scale bioreactor:
Application to in vivo NMR measurement. Biotechnology and Bioengineering, 89, 138-
147.
Goff, K. L., L. Quaroni & K. E. Wilson (2009) Measurement of metabolite formation in single
living cells of Chlamydomonas reinhardtii using synchrotron Fourier-Transform
Infrared spectromicroscopy. Analyst, 134, 2216-2219.
Graf, T. & M. Stadtfeld (2008) Heterogeneity of Embryonic and Adult Stem Cells. Cell Stem
Cell, 3, 480-483.
Grivet, J.-P. & A.-M. Delort (2009) NMR for microbiology: In vivo and in situ applications.
Progress in Nuclear Magnetic Resonance Spectroscopy, 54, 1-53.
Computational Methods to Interpret and Integrate Metabolomic Data 123

Gu, H., H. Chen, Z. Pan, A. U. Jackson, N. Talaty, B. Xi, C. Kissinger, C. Duda, D. Mann, D.
Raftery & R. G. Cooks (2007) Monitoring diet effects via biofluids and their
implications for metabolomics studies. Analytical Chemistry, 79, 89-97.
Guyon, I., J. Weston, S. Barnhill & V. Vapnik (2002) Gene Selection for Cancer Classification
using Support Vector Machines. Machine Learning, 46, 389-422.
Gygi, S. P., Y. Rochon, B. R. Franza & R. Aebersold (1999) Correlation between protein and
mRNA abundance in yeast. Molecular and Cellular Biology, 19, 1720-30.
Hageman, J. A., R. A. van den Berg, J. A. Westerhuis, H. C. J. Hoefsloot & A. K. Smilde
(2006) Bagged K-Means Clustering of Metabolome Data. Critical Reviews in
Analytical Chemistry, 36, 211-220.
Heinemann, M. & R. Zenobi (2011) Single cell metabolomics. Current Opinion in
Biotechnology. 22, 26-31.
Heinrich, R. & S. Schuster. 1996. The Regulation of Cellular Systems. Chapman and Hall.
Hermelink, A., A. Brauer, P. Lasch & D. Naumann (2009) Phenotypic heterogeneity within
microbial populations at the single-cell level investigated by confocal Raman
microspectroscopy. Analyst, 134, 1149-1153.
Hirai, M. Y., M. Klein, Y. Fujikawa, M. Yano, D. B. Goodenowe, Y. Yamazaki, S. Kanaya, Y.
Nakamura, M. Kitayama, H. Suzuki, N. Sakurai, D. Shibata, J. Tokuhisa, M.
Reichelt, J. Gershenzon, J. Papenbrock & K. Saito (2005) Elucidation of gene-to-gene
and metabolite-to-gene networks in arabidopsis by integration of metabolomics
and transcriptomics. Journal of Biological Chemistry, 280, 25590-5.
Hirai, M. Y., M. Yano, D. B. Goodenowe, S. Kanaya, T. Kimura, M. Awazuhara, M. Arita, T.
Fujiwara & K. Saito (2004) Integration of transcriptomics and metabolomics for
understanding of global responses to nutritional stresses in Arabidopsis thaliana.
Proceedings of the National Academy of Sciences of the United States of America, 101,
10205-10.
Hollywood, K., D. R. Brison & R. Goodacre (2006) Metabolomics: Current technologies and
future trends. Proteomics, 6, 4716-4723.
Holmes, D., D. Pettigrew, C. H. Reccius, J. D. Gwyer, C. van Berkel, J. Holloway, D. E.
Davies & H. Morgan (2009) Leukocyte analysis and differentiation using high
speed microfluidic single cell impedance cytometry. Lab on a Chip, 9, 2881-2889.
Hoops, S., S. Sahle, R. Gauges, C. Lee, J. Pahle, N. Simus, M. Singhal, L. Xu, P. Mendes & U.
Kummer. 2006. COPASI--a COmplex PAthway SImulator. In Bioinformatics, 3067-
74. England.
Hsieh, Y., R. Casale, E. Fukuda, J. W. Chen, I. Knemeyer, J. Wingate, R. Morrison & W.
Korfmacher (2006) Matrix-assisted laser desorption/ionization imaging mass
spectrometry for direct measurement of clozapine in rat brain tissue. Rapid
Communications in Mass Spectrometry, 20, 965-972.
Huang, L. & R. T. Kennedy (1995) Exploring single-cell dynamics using chemically-modified
microelectrodes. Trends in Analytical Chemistry, 14, 158-164.
Irish, J. M., N. Kotecha & G. P. Nolan (2006) Innovation - Mapping normal and cancer cell
signalling networks: towards single-cell proteomics. Nature Reviews Cancer, 6, 146-
155.
Ishii, N. & M. Tomita. 2009. Multi-Omics Data-Driven Systems Biology of E. coli.
Jones, J. J., S. Borgmann, C. L. Wilkins & R. M. O'Brien (2006) Characterizing the
phospholipid profiles in mammalian tissues by MALDI FTMS. Analytical Chemistry,
78, 3062-3071.
124 Metabolomics

Jewett, M., M. Hansen & J. Nielsen. 2007a. Data acquisition, analysis, and mining:
Integrative tools for discerning. In Metabolomics: A Powerful Tool in Systems Biology,
eds. J. Nielsen & M. C. Jewett, 159-187. Springer.
---. 2007b. Data acquisition, analysis, and mining: Integrative tools for discerning. In
Metabolomics, A Powerful Tool in Systems Biology, eds. J. Nielsen & M. C. Jewett, 159-
187. Springer.
Jonsson, P., S. J. Bruce, T. Moritz, J. Trygg, M. Sjostrom, R. Plumb, J. Granger, E. Maibaum, J.
K. Nicholson, E. Holmes & H. Antti (2005) Extraction, interpretation and validation
of information for comparing samples in metabolic LC/MS data sets. Analyst, 130,
701-7.
Kaartinen, J., Y. Hiltunen, P. T. Kovanen & M. Ala-Korpela (1998) Application of self-
organizing maps for the detection and classification of human blood plasma
lipoprotein lipid profiles on the basis of 1H NMR spectroscopy data. NMR in
Biomedicine, 11, 168-176.
Kandpal, R. P., B. Saviola & J. Felton (2009) The era of 'omics unlimited. Biotechniques, 46,
351.
Kanaya, S., M. Kinouchi, T. Abe, Y. Kudo, Y. Yamada, T. Nishi, H. Mori & T. Ikemura (2001)
Analysis of codon usage diversity of bacterial genes with a self-organizing map
(SOM): characterization of horizontally transferred genes with emphasis on the E.
coli O157 genome. Gene, 276, 89-99.
Khatib-Shahidi, S., M. Andersson, J. L. Herman, T. A. Gillespie & R. M. Caprioli (2006)
Direct molecular analysis of whole-body animal tissue sections by imaging MALDI
mass spectrometry. Analytical Chemistry, 78, 6448-6456.
Kind, T. & O. Fiehn. 2006. Metabolomic database annotations via query of elemental
compositions: mass accuracy is insufficient even at less than 1 ppm. In BMC
Bioinformatics, 234 7:234. England.
Klamt, S. & E. D. Gilles (2004) Minimal cut sets in biochemical reaction networks.
Bioinformatics, 20, 226-34.
Klamt, S., J. Saez-Rodriguez & E. D. Gilles (2007) Structural and functional analysis of
cellular networks with CellNetAnalyzer. BMC Systems Biology, 1, 2.
Klamt, S., J. Saez-Rodriguez, J. A. Lindquist, L. Simeoni & E. D. Gilles (2006) A methodology
for the structural and functional analysis of signaling and regulatory networks.
BMC Bioinformatics, 7, 56.
Klamt, S. & S. Schuster (2002) Calculating as many fluxes as possible in underdetermined
metabolic networks. Mol Biol Rep, 29, 243-8.
Klamt, S. & J. Stelling (2003) Two approaches for metabolic pathway analysis? Trends in
Biotechnology, 21, 64-9.
Kohonen, T. (1998) The self-organizing map. Neurocomputing, 21, 1-6.
---. 2001. Self-Organizing Maps. Springer.
Kouskoumvekaki, I., Z. Yang, S. O. Jonsdottir, L. Olsson & G. Panagiotou. 2008.
Identification of biomarkers for genotyping Aspergilli using non-linear methods for
clustering and classification. In BMC Bioinformatics, 59. England.
Kummel, A., S. Panke & M. Heinemann (2006) Putative regulatory sites unraveled by
network-embedded thermodynamic analysis of metabolome data. Molecular
Systems Biology, 2, 2006 0034.
Le Cao, K. A., P. G. Martin, C. Robert-Granie & P. Besse (2009) Sparse canonical methods for
biological data integration: application to a cross-platform study. BMC
Bioinformatics, 10, 34.
Computational Methods to Interpret and Integrate Metabolomic Data 125

Le Cao, K. A., D. Rossouw, C. Robert-Granie & P. Besse (2008) A sparse PLS for variable
selection when integrating omics data. Statistical and Applied Genetics and Molecular
Biology, 7, Article 35.
Lenz, E. M. & I. D. Wilson (2007) Analytical strategies in metabonomics. Journal of Proteome
Research, 6, 443-458.
Lei, F. & S. B. Jorgensen. 2001. Estimation of kinetic parameters in a structured yeast model
using regularisation. In Journal of Biotechnology, 223-37. Netherlands.
Lerouxel, O., T. S. Choo, M. Seveno, B. Usadel, L. Faye, P. Lerouge & M. Pauly (2002) Rapid
structural phenotyping of plant cell wall mutants by enzymatic oligosaccharide
fingerprinting. Plant Physiology, 130, 1754-1763.
Li, X., X. Lu, J. Tian, P. Gao, H. Kong & G. Xu (2009) Application of fuzzy c-means clustering
in data analysis of metabolomics. Analytical Chemistry, 81, 4468-75.
Lin, J. & J. Qian (2007) Systems biology approach to integrative comparative genomics.
Expert Review of Proteomics, 4, 107-119.
Lindon, J. C., E. Holmes & J. K. Nicholson (2003) So whats the deal with metabonomics?
Metabonomics measures the fingerprint of biochemical perturbations caused by
disease, drugs, and toxins. Analytical Chemistry, 75, 384A-391A.
Lindon, J. C., J. K. Nicholson & I. D. Wilson (2000) Directly coupled HPLC-NMR and HPLC-
NMR-MS in pharmaceutical research and development. Journal of Chromatography
B, 748, 233-258.
Listgarten, J. & A. Emili. 2005. Statistical and computational methods for comparative
proteomic profiling using liquid chromatography-tandem mass spectrometry. In
Molecular and Cellular Proteomics, 419-34. United States.
Lu, K.-Y., A. M. Wo, Y.-J. Lo, K.-C. Chen, C.-M. Lin & C.-R. Yang (2006) Three dimensional
electrode array for cell lysis via electroporation. Biosensors & Bioelectronics, 22, 568-
574.
Mahadevan, S., S. L. Shah, T. J. Marrie & C. M. Slupsky (2008) Analysis of metabolomic data
using support vector machines. Analytical Chemistry, 80, 7562-70.
Mallouchos, A., M. Komaitis, A. Koutinas & M. Kanellaki (2002) Investigation of volatiles
evolution during the alcoholic fermentation of grape must using free and
immobilized cells with the help of solid phase microextraction (SPME) headspace
sampling. Journal of Agricultural and Food Chemistry, 50, 3840-3848.
Mas, S., S. G. Villas-Boas, M. E. Hansen, M. Akesson & J. Nielsen (2007) A comparison of
direct infusion MS and GC-MS for metabolic footprinting of yeast mutants.
Biotechnology and Bioengineering, 96, 1014-1022.
Masujima, T. (2009) Live single-cell mass spectrometry. Analytical Sciences, 25, 953-960.
Mattoli, L., F. Cangi, A. Maidecchi, C. Ghiara, E. Ragazzi, M. Tubaro, L. Stella, F. Tisato & P.
Traldi (2006) Metabolomic fingerprinting of plant extracts. Journal of Mass
Spectrometry, 41, 1534-1545.
Maxwell, R. J., I. Martinez-Perez, S. Cerdan, M. E. Cabanas, C. Arus, A. Moreno, A.
Capdevila, E. Ferrer, F. Bartomeus, A. Aparicio, G. Conesa, J. M. Roda, F. Carceller,
J. M. Pascual, S. L. Howells, R. Mazucco & J. R. Griffiths (1998) Pattern recognition
analysis of 1H NMR spectra from perchloric acid extracts of human brain tumor
biopsies. Magnetic Resonance in Medicine, 39, 869-77.
Mellors, J. S., V. Gorbounov, R. S. Ramsey & J. M. Ramsey (2008) Fully integrated glass
microfluidic device for performing high-efficiency capillary electrophoresis and
electrospray ionization mass spectrometry. Analytical Chemistry, 80, 6881-6887.
126 Metabolomics

Meija, J. (2006) Mathematical tools in analytical mass spectrometry. Analytical and


Bioanalytical Chemistry, 486-499.
Meinicke, P., T. Lingner, A. Kaever, K. Feussner, C. Gobel, I. Feussner, P. Karlovsky & B.
Morgenstern. 2008. Metabolite-based clustering and visualization of mass
spectrometry data using one-dimensional self-organizing maps. In Algorithms Mol
Biol, 9. England.
Moles, C. G., P. Mendes & J. R. Banga (2003) Parameter Estimation in Biochemical Pathways:
A Comparison of Global Optimization Methods. Genome Research, 13, 2467-2474.
Monton, M. R. N. & T. Soga (2007) Metabolome analysis by capillary electrophoresis-mass
spectrometry. Journal of Chromatography A, 1168, 237-246.
Motta, A., D. Paris & D. Melck (2010) Monitoring real-time metabolism of living cells by fast
two-dimensional NMR spectroscopy. Analytical Chemistry, 82, 2405-2411.
Mylonas, R., Y. Mauron, A. Masselot, P. A. Binz, N. Budin, M. Fathi, V. Viette, D. F.
Hochstrasser & F. Lisacek (2009) X-Rank: a robust algorithm for small molecule
identification using tandem mass spectrometry. Analytical Chemistry, 81, 7604-10.
Neumann, S. & S. Bocker (2010) Computational mass spectrometry for metabolomics:
Identification of metabolites and small molecules. Analytical and Bioanalytical
Chemistry, 2779-2788.
Noteborn, H., A. Lommen, R. C. van der Jagt & J. M. Weseman (2000) Chemical
fingerprinting for the evaluation of unintended secondary metabolic changes in
transgenic food crops. Journal of Biotechnology, 77, 103-114.
Oberacher, H., M. Pavlic, K. Libiseller, B. Schubert, M. Sulyok, R. Schuhmacher, E. Csaszar
& H. C. Kofeler (2009) On the inter-instrument and the inter-laboratory
transferability of a tandem mass spectral reference library: 2. Optimization and
characterization of the search algorithm. Journal of Mass Spectrometry, 44, 494-502.
Ott, K. H., N. Aranibar, B. Singh & G. W. Stockton. 2003. Metabonomics classifies pathways
affected by bioactive compounds. Artificial neural network classification of NMR
spectra of plant extracts. In Phytochemistry, 971-85. United States.
Papin, J. A., J. Stelling, N. D. Price, S. Klamt, S. Schuster & B. O. Palsson. 2004. Comparison
of network-based pathway analysis methods. In Trends in Biotechnology, 400-5.
England.
Pir, P., B. Kirdar, A. Hayes, Z. Y. Onsan, K. O. Ulgen & S. G. Oliver (2006) Integrative
investigation of metabolic and transcriptomic data. BMC Bioinformatics, 7, 203.
Podwojski, K., A. Fritsch, D. C. Chamrad, W. Paul, B. Sitek, P. Mutzel, C. Stephan, H. E.
Meyer, W. Urfer, K. Ickstadt & J. Rahnenführer (2009) Retention Time Alignment
Algorithms for LC/MS Data must consider Nonlinear Shifts. Bioinformatics. 25: 758-
64.
Pohjanen, E., E. Thysell, J. Lindberg, I. Schuppe-Koistinen, T. Moritz, P. Jonsson & H. Antti
(2006) Statistical multivariate metabolite profiling for aiding biomarker pattern
detection and mechanistic interpretations in GC/MS based metabolomics.
Metabolomics, 2, 257-268-268.
Price, N. D., J. A. Papin, C. H. Schilling & B. O. Palsson (2003) Genome-scale microbial in
silico models: the constraints-based approach. Trends in Biotechnology, 21, 162-9.
Raamsdonk, L. M., B. Teusink, D. Broadhurst, N. S. Zhang, A. Hayes, M. C. Walsh, J. A.
Berden, K. M. Brindle, D. B. Kell, J. J. Rowland, H. V. Westerhoff, K. van Dam & S.
G. Oliver (2001) A functional genomics strategy that uses metabolome data to
reveal the phenotype of silent mutations. Nature Biotechnology, 19, 45-50.
Computational Methods to Interpret and Integrate Metabolomic Data 127

Ramautar, R., A. Demirci & G. J. de Jong (2006) Capillary electrophoresis in metabolomics.


Trac-Trends in Analytical Chemistry, 25, 455-466.
Rashed, M. S., M. P. Bucknall, D. Little, A. Awad, M. Jacob, M. Alamoudi, M. Alwattar & P.
T. Ozand (1997) Screening blood spots for inborn errors of metabolism by
electrospray tandem mass spectrometry with a microplate batch process and a
computer algorithm for automated flagging of abnormal profiles. Clinical Chemistry,
43, 1129-1141.
Reiner, A., D. Yekutieli & Y. Benjamini (2003) Identifying differentially expressed genes
using false discovery rate controlling procedures. Bioinformatics, 19, 368-75.
Reyzer, M. L. & R. M. Caprioli (2007) MALDI-MS-based imaging of small molecules and
proteins in tissues. Current Opinion in Chemical Biology, 11, 29-35.
Roessner, U., C. Wagner, J. Kopka, R. N. Trethewey & L. Willmitzer (2000) Simultaneous
analysis of metabolites in potato tuber by gas chromatography-mass spectrometry.
Plant Journal, 23, 131-142.
Roessner-Tunali, U., B. Hegemann, A. Lytovchenko, F. Carrari, C. Bruedigam, D. Granot &
A. R. Fernie (2003) Metabolic profiling of transgenic tomato plants overexpressing
hexokinase reveals that the influence of hexose phosphorylation diminishes during
fruit development. Plant Physiology, 133, 84-99.
Rubakhin, S. S., J. C. Jurchen, E. B. Monroe & J. V. Sweedler (2005) Imaging mass
spectrometry: fundamentals and applications to drug discovery. Drug Discovery
Today, 10, 823-837.
Rubingh, C. M., S. Bijlsma, R. H. Jellema, K. M. Overkamp, M. J. van der Werf & A. K.
Smilde (2009) Analyzing longitudinal microbial metabolomics data. Journal of
Proteome Research, 8, 4319-27.
Sato, S., T. Soga, T. Nishioka & M. Tomita (2004) Simultaneous determination of the main
metabolites in rice leaves using capillary electrophoresis mass spectrometry and
capillary electrophoresis diode array detection. Plant Journal, 40, 151-163.
Schmid, A., H. Kortmann, P. S. Dittrich & L. M. Blank (2010) Chemical and biological single
cell analysis. Current Opinion in Biotechnology, 21, 12-20.
Schilling, C. H., J. S. Edwards, D. Letscher & B. Palsson (2000a) Combining pathway analysis
with flux balance analysis for the comprehensive study of metabolic systems.
Biotechnology & Bioengineering, 71, 286-306.
Schilling, C. H., D. Letscher & B. O. Palsson (2000b) Theory for the systemic definition of
metabolic pathways and their use in interpreting metabolic function from a
pathway-oriented perspective. Journal of Theoretical Biology, 203, 229-48.
Schilling, C. H., S. Schuster, B. O. Palsson & R. Heinrich (1999) Metabolic pathway analysis:
basic concepts and scientific applications in the post-genomic era. Biotechnology
Progress, 15, 296-303.
Schrader, B., H. H. Klump, K. Schenzel & H. Schulz (1999) Non-destructive NIR FT Raman
analysis of plants. Journal of Molecular Structure, 509, 201-212.
Schroer, K., B. Zelic, M. Oldiges & S. Lutz (2009) Metabolomics for biotransformations:
Intracellular redox cofactor analysis and enzyme kinetics offer insight into whole
cell processes. Biotechnology & Bioengineering, 104, 251-60.
Schuster, K. C., I. Reese, E. Urlaub, J. R. Gapes & B. Lendl (2000) Multidimensional
information on the chemical composition of single bacterial cells by confocal
Raman microspectroscopy. Analytical Chemistry, 72, 5529-5534.
128 Metabolomics

Schuster, S., T. Pfeiffer, F. Moldenhauer, I. Koch & T. Dandekar (2002) Exploring the
pathway structure of metabolism: decomposition into subnetworks and application
to Mycoplasma pneumoniae. Bioinformatics, 18, 351-361.
Seger, C. & S. Sturm (2007) Analytical aspects of plant metabolite profiling platforms:
Current standings and future aims. Journal of Proteome Research, 6, 480-497.
Segre, D., J. Zucker, J. Katz, X. Lin, P. D'Haeseleer, W. P. Rindone, P. Kharchenko, D. H.
Nguyen, M. A. Wright & G. M. Church (2003) From annotated genomes to
metabolic flux models and kinetic parameter fitting. OMICS, 7, 301-16.
Shinohara, H. & F. Wang (2007) Real-time detection of dopamine released from a nerve
model cell by an enzyme-catalyzed luminescence method and its application to
drug assessment. Analytical Sciences, 23, 81-84.
Singh, O. V. & N. S. Nagaraj (2006) Transcriptomics, proteomics and interactomics: unique
approaches to track the insights of bioremediation. Briefings in Functional Genomics
& Proteomics, 4, 355-362.
Soga, T., Y. Ohashi, Y. Ueno, H. Naraoka, M. Tomita & T. Nishioka (2003) Quantitative
metabolome analysis using capillary electrophoresis mass spectrometry. Journal of
Proteome Research, 2, 488-494.
Southam, A. D., T. G. Payne, H. J. Cooper, T. N. Arvanitis & M. R. Viant (2007) Dynamic
range and mass accuracy of wide-scan direct infusion nanoelectrospray
Fourier transform ion cyclotron resonance mass spectrometry-based metabolomics
increased by the spectral stitching method. Analytical Chemistry, 79, 4595-4602.
Stein, S. E. (1994) Estimating probabilities of correct identification from results of mass
spectral library searches. Journal of the American Society for Mass Spectrometry, 5, 316-
323.
Stelling, J., S. Klamt, K. Bettenbrock, S. Schuster & E. D. Gilles (2002) Metabolic network
structure determines key aspects of functionality and regulation. Nature, 420, 190-
193.
Stephanopoulos, G., H. Alper & J. Moxley (2004) Exploiting biological complexity for strain
improvement through systems biology. Nature Biotechnology, 22, 1261-1267.
Steuer, R. 2006. Review: on the analysis and interpretation of correlations in metabolomic
data. In Briefing in Bioinformatics, 151-8. England.
---. 2007. Computational approaches to the topology, stability and dynamics of metabolic
networks. In Phytochemistry, 2139-51. United States.
Storey, J. D. (2002) A direct approach to false discovery rates. Journal of the Royal Statistical
Society: Series B (Statistical Methodology), 64, 479-498.
Storey, J. D. & R. Tibshirani. 2003. Statistical significance for genomewide studies. In
Proceedings of the National Academy of Sciences of the United States of America, 9440-5.
United States.
Strelkov, S., M. von Elstermann & D. Schomburg (2004) Comprehensive analysis of
metabolites in Corynebacterium glutamicum by gas chromatography/mass
spectrometry. Biological Chemistry, 385, 853-861.
Sturm, S., C. Seger & H. Stuppner (2007) Analysis of Central European Corydalis species by
nonaqueous capillary electrophoresis-electrospray ion trap mass spectrometry.
Journal of Chromatography A, 1159, 42-50.
Suits, F., J. Lepre, P. Du, R. Bischoff & P. Horvatovich (2008) Two-dimensional method for
time aligning liquid chromatography-mass spectrometry data. Analytical Chemistry,
80, 3095-104.
Computational Methods to Interpret and Integrate Metabolomic Data 129

Sumner, L., A. Amberg, D. Barrett, M. Beale, R. Beger, C. Daykin, T. Fan, O. Fiehn, R.


Goodacre, J. Griffin, T. Hankemeier, N. Hardy, J. Harnly, R. Higashi, J. Kopka, A.
Lane, J. Lindon, P. Marriott, A. Nicholls, M. Reily, J. Thaden & M. Viant (2007)
Proposed minimum reporting standards for chemical analysis. Metabolomics, 211-
221.
Sysi-Aho, M., M. Katajamaa, L. Yetukuri & M. Oresic. 2007. Normalization method for
metabolomics data using optimal selection of multiple internal standards. In BMC
Bioinformatics, 8: 93. England.
Takats, Z., J. M. Wiseman, B. Gologan & R. G. Cooks (2004) Mass spectrometry sampling
under ambient conditions with desorption electrospray ionization. Science, 306, 471-
473.
Talaty, N., Z. Takats & R. G. Cooks (2005) Rapid in situ detection of alkaloids in plant tissue
under ambient conditions using desorption electrospray ionization. Analyst, 130,
1624-1633.
Tanaka, K., A. Westdull, D. G. Hine, T. B. Lynn & T. Lowe (1980) Gas-chromategraphic
method of analysis for urinary organic-acids. 2. Description of the procedure, and
its application to diagnosis of patients with organic acidurias. Clinical Chemistry, 26,
1847-1853.
Tapp, H. S. & E. K. Kemsley (2009) Notes on the practical utility of OPLS. TrAC Trends in
Analytical Chemistry, 28, 1322-1327.
Tibshirani, R. (1996) Regression shrinkage and selection via the Lasso. Journal of the Royal
Statistical Society Series B-Methodological, 58, 267-288.
Trygg, J. & T. Lundstedt. 2007. Chemometrics Techniques for Metabonomics. In The
Handbook of Metabonomics and Metabolomics, eds. J. C. Lindon, J. K. Nicholson & E.
Holmes. London: Elseivier.
Urban, P. L., K. Jefimovs, A. Amantonico, S. R. Fagerer, T. Schmid, S. Maedler, J. Puigmarti-
Luis, N. Goedecke & R. Zenobi (2010) High-density micro-arrays for mass
spectrometry. Lab on a Chip, 10, 3206-3209.
Urbanczik, R. & C. Wagner (2005) An improved algorithm for stoichiometric network
analysis: theory and applications. Bioinformatics, 21, 1203-10.
Urbanczyk-Wochniak, E., A. Luedemann, J. Kopka, J. Selbig, U. Roessner-Tunali, L.
Willmitzer & A. R. Fernie (2003) Parallel analysis of transcript and metabolic
profiles: a new approach in systems biology. EMBO Report, 4, 989-93.
Urbanczyk-Wochniak, E., L. Willmitzer & A. R. Fernie (2007) Integrating profiling data:
using linear correlation to reveal coregulation of transcript and metabolites.
Methods in Molecular Biology, 358, 77-85.
van den Berg, R. A., C. M. Rubingh, J. A. Westerhuis, M. J. van der Werf & A. K. Smilde
(2009) Metabolomics data exploration guided by prior knowledge. Anal Chim Acta,
651, 173-81.
van Nederkassel, A. M., M. Daszykowski, P. H. Eilers & Y. V. Heyden. 2006. A comparison
of three algorithms for chromatograms alignment. In J Chromatogr A, 199-210.
Netherlands.
van Velzen, E. J., J. A. Westerhuis, J. P. van Duynhoven, F. A. van Dorsten, C. H. Grun, D.
M. Jacobs, G. S. Duchateau, D. J. Vis & A. K. Smilde (2009) Phenotyping tea
consumers by nutrikinetic analysis of polyphenolic end-metabolites. Journal of
Proteome Research, 8, 3317-30.
130 Metabolomics

Vandenbogaert, M., S. Li-Thiao-Te, H. M. Kaltenbach, R. Zhang, T. Aittokallio & B.


Schwikowski (2008) Alignment of LC-MS images, with applications to biomarker
discovery and protein identification. Proteomics, 8, 650-72.
Varma, A. & B. O. Palsson (1994) Metabolic Flux Balancing: Basic Concepts, Scientific and
Practical Use. Nature Biotechnology, 12, 994-998.
Villas-Boas, S. G., S. Mas, M. Akesson, J. Smedsgaard & J. Nielsen (2005) Mass spectrometry
in metabolome analysis. Mass Spectrometry Reviews, 24, 613-646.
Visser, D. & J. J. Heijnen. 2003. Dynamic simulation and metabolic re-design of a branched
pathway using linlog kinetics. In Metabolic Engineering, 164-76. United States.
Wagner, C., M. Sefkow & J. Kopka (2003) Construction and application of a mass spectral
and retention time index database generated from plant GC/EI-TOF-MS metabolite
profiles. Phytochemistry, 62, 887-900.
Wang, D. & S. Bodovitz (2010) Single cell analysis: the new frontier in 'omics'. Trends in
Biotechnology, 28, 281-290.
Warrack, B. M., S. Hnatyshyn, K. H. Ott, M. D. Reily, M. Sanders, H. Zhang & D. M. Drexler.
2009. Normalization strategies for metabonomic analysis of urine samples. In J
Chromatogr B Analyt Technol Biomed Life Sci, 547-52. Netherlands.
Webb-Robertson, B. J., L. A. McCue, N. Beagley, J. E. McDermott, D. S. Wunschel, S. M.
Varnum, J. Z. Hu, N. G. Isern, G. W. Buchko, K. McAteer, J. G. Pounds, S. J.
Skerrett, D. Liggitt & C. W. Frevert (2009) A Bayesian integration model of high-
throughput proteomics and metabolomics data for improved early detection of
microbial infections. Pac Symp Biocomput, 451-63.
Weckwerth, W., M. E. Loureiro, K. Wenzel & O. Fiehn (2004) Differential metabolic
networks unravel the effects of silent plant phenotypes. Proceedings of the National
Academy of Sciences of the United States of America, 101, 7809-7814.
Weckwerth, W., K. Wenzel & O. Fiehn (2004) Process for the integrated extraction,
identification and quantification of metabolites, proteins and RNA to reveal their
co-regulation in biochemical networks. Proteomics, 4, 78-83.
Wiseman, J. M., S. M. Puolitaival, Z. Takats, R. G. Cooks & R. M. Caprioli (2005) Mass
spectrometric profiling of intact biological tissue by using desorption electrospray
ionization. Angewandte Chemie-International Edition, 44, 7094-7097.
Wittmann, C., J. O. Kromer, P. Kiefer, T. Binz & E. Heinzle (2004) Impact of the cold shock
phenomenon on quantification of intracellular metabolites in bacteria. Analytical
Biochemistry, 327, 135-139.
Xie, Y., W. Pan & A. B. Khodursky (2005) A note on using permutation-based false discovery
rate estimates to compare different analysis methods for microarray data.
Bioinformatics, 21, 4280-4288.
Yassaa, N., E. Brancaleoni, M. Frattoni & P. Ciccioli (2001) Trace level determination of
enantiomeric monoterpenes in terrestrial plant emission and in the atmosphere
using a beta-cyclodextrin capillary column coupled with thermal desorption and
mass spectrometry. Journal of Chromatography A, 915, 185-197.
Zhang, J., E. Gonzalez, T. Hestilow, W. Haskins & Y. Huang (2009) Review of peak detection
algorithms in liquid-chromatography-mass spectrometry. Current Genomics, 10,
388-401.
Zhang, W., F. Li & L. Nie (2010) Integrating multiple 'omics' analysis for microbial biology:
application and methodologies. Microbiology, 156, 287-301.
6

Metabotype Concept: Flexibility, Usefulness and


Meaning in Different Biological Populations
Nabil Semmar1, 2
1Plateau BioMeT, UMR INRA 1260, Medical School of Marseilles,Marseille,
2Université de Tunis El Manar, Institut Supérieur des Sciences Biologiques Appliquées de
Tunis (ISSBAT), Rue Zouhair Essafi, Tunis,
1France
2Tunisia

1. Introduction
Metabolism represents a junction system in biological body receiving cumulated signals
from upstream (genome, transcriptome, proteome) and downstream (environment) systems.
This median position of the metabolic system makes it to be very sensitive toward internal
and external signals resulting in its regulatory role in physiological homeostasy and
adaptive responses to endogenous and exogenous factors. These factors have initiation,
modulation or pressure effects on the biological organisms and species which adapt, resist
or react through different types of metabolisms based on different synthesis and regulation
levels of metabolites (Wilson, 2009).
These characteristics give to metabolism a flexibility that may be described by means of four
variability criteria (Fig. 1a): presence-absence of metabolites, concentration levels, relative
levels or ratios between metabolites, and metabolic profiles characterizing different structural
and functional states in biosystems through different metabolites’ levels. Presence-absence of
metabolites is a qualitative criterion that concerns metabolites that are stimulated by particular
internal biological states (species physiology, disease, stress, etc.) or external governing factors
(climate, threat, etc.). Beyond this binary aspect of metabolic responses, increase or decrease in
concentration levels of some metabolites can be sensitive responses to the degree of a
governing implicit factor (e.g. wounding, toxin, pollutant exposure levels, etc.). More precision
on association between biological state and governing factor can be extracted from
concentration ratios between sensitive metabolites. Complex situations integrating many
interactive factors can be characterized by metabolic profiles in which several metabolites
levels increase or decrease compared with control conditions or neutral situations.
The four metabolic variability criteria can be used separately or in association to provide reliable
pictures on different metabolic phenotypes of a biological system; such pictures characterizing
the metabolic phenotypes are called metabotypes (or chemotypes) (Fig. 1a). Identifications of
relationships between metabotypes and quantitative or qualitative control factors lead to the
concept of metabolic markers. Metabolic markers can be used to anticipate (predict), alert and
control responses and states of different components in biological systems. These components
include cells, biofluids, tissues, organisms and biological populations or species.
132 Metabolomics

Presence- Concentration
Absence levels
A B

C D C D Concentrations
(a) (µg/L)
AC AD BC BD
Major metabolite
A
Metabolites B
A, B, C, D C
D Minor

    Presence Absence

Metabotypes
Metabolite B
40% B 80% B 100% B Concentrations or
relative levels

100% 60% A 20% A


ABCD ABCD ABCD
Metabolite A
Metabolites

Relative levels
(ratios) Profiling

 Gender, age, race identification  Dietary sources classification


(b)
 Biorythmic processes analysis  Intake amount estimation
 Obesity-linked responses analysis  Metabolizer types identification

Physiology Nutrition

METABOTYPE CRITERIA:

- Presence-Absence of Metabolites
- Concentration Levels
- Ratios between Metabolites’ levels
- Profiling of Metabolites’ sets

Ecology Clinics

 Environmental conditions deduction  Diseases determination


 Biological signals interpretation  Intoxication sources identification
 Geographic origins control  Doping detection
 Chemotaxonomical analysis

Fig. 1. (a) Schematic representations of four metabotype definitions based on occurrences,


concentration levels, ratios and profiles of metabolites in biological matrices. (b) Interest of
the four metabotype criteria to develop biomarkers in different biological fields (physiology,
nutrition, clinics, ecology).
Metabotype Concept: Flexibility, Usefulness and Meaning in Different Biological Populations 133

This chapter illustrates the usefulness of metabotype concept as flexible biomarkers in


different biological fields: physiology, clinics, nutrition and ecology (Fig. 1b): in physiology,
metabotypes can be markers of biological varieties, gender, aging, biorhythms, etc. (Bell et
al., 1991; Bollard et al., 2001; Duncan et al., 2007 ; Holmes et al., 2008). In nutrition, different
metabotypes can be correlated to different diets; secondary metabolites, particularly
produced by plants, represent strong biomarkers of consumed fruits, herbals and legumes
(Kaufman et al., 1997; Yanez, 2008). In clinical field, different metabotypes can be indicator
of different disease types, intoxication levels or doping sources (Timbrell, 1998; Kintz et al.,
1999; Barderas et al., 2011). In ecology, different plant metabotypes based on secondary
metabolites are indicative of different physicochemical conditions (temperature, light,
humidity, oxidative stress, etc.) or biological relationships (parasite or herbivore attacks,
pollination states, etc.) submitted by the plant (Semmar et al., 2008). Also, metabotype (or
chemotype) concept is helpfully used in chemotaxonomy to highlight chemical
polymorphism such as chemical varieties of floral colors (Torskangerpoll et al., 2005).

2. Physiological metabotypes
2.1 Metabotypes based on metabolites’ occurrences
Presence-absence of metabolites is an efficient metabolomic parameter to characterize
different biological species or varieties known to represent different physiological systems
(Fig. 2): for instance, benzoic acid is metabolized almost entirely to hippuric acid by
primates, rodents and rabbits (mammals) (Fig. 2a). However, it is excreted unchanged and
as glucuronide by insects, birds and reptiles (Jones, 1982). Similarly, the excretion of
phenylacetic acid (Fig. 2b) as the parent compound or as glutamine, glycine or taurine
conjugates is species-dependent (Robertson et al., 2002): for instance, it is excreted as
phenylacetyl glutamine in humans and phenylacetylglycine in rats.
Comparison of 1H NMR spectra from control B6C3F1 mouse urines with those of control SD
rat urines revealed the presence of guanidinoacetic acid and trimethylamine in mouse but
their absence in rat (Bollard et al., 2005).

2.2 Metabotypes based on concentration levels


In animals, several metabolites have been found to quantitatively vary in relation to gender,
age, body weight, etc. :
Plasma NMR profiles showed reduced concentration of the triglyceride resonances in
female rats compared with male counterparts (Fig.3a) (Stanley, 2002).
In humans, urinary citrate levels have been found to be generally greater in females (Fig.3b)
(Hodgkinson, 1962). Moreover, serum cholesterol levels are different between sexes at
matched ages, with in general, pre-menopausal females being less susceptible to high
cholesterol levels than males (Fig.3c) (Joossen, 1988).
Oestrogenic hormones are known to exhibit some control over the kidney: in humans,
glycosuria (glucose in urine) is common in pregnant females (Fig.3d) (Davison and Hytten,
1975). In the rat, pregnancy is associated with an increased glucose filtration rate and
decreased urine flow rate (Bishop and Green, 1980).
134 Metabolomics

COOH
(a)

Benzoic acid

OH Urinary excretion
OH
N
O
Hippuric acid - Benzoic acid
O
- Glucuronyl Benzoic acid OH OH
OH
O O
HOOC

Primates Insects
Rodents Birds
Rabbits Reptiles

OH
(b)
O

Phenylacetic acid

O Urinary excretion O

NH NH
OH
OH
O O
Phenylacetylglutamine Phenylacetylglycine
H2N O

Humans Rats

Fig. 2. Metabolic derivatives of benzoic acid (a) and phenylacetic acid (b) excreted in urine of
human and animal species and from which specific metabotypes can be defined.
Metabotype Concept: Flexibility, Usefulness and Meaning in Different Biological Populations 135

(a) Plasm a triglycerides


(b)
Urinary citrate
Indicative levels
H 2C O CO R1
R1, R2, R3: O OH
HC O CO R2 O O
Fatty acids
H 2C O CO R3
Higher HO OH
OH

Lower

Male rats Fem ale Male Fem ale


rats hum ans hum ans

(c) Serum H3C CH3 (d) (e)


Cholesterol CH3 Urinary glucose Fecal propanoic acid
CH3 OH
CH3 O
HO O
OH H3C CH 2 C
HO
OH
HO OH

Male Pre-menopausal Male pregnant Lean Obese


hum ans fem ale humans hum ans fem ale humans hum ans hum ans

(g) (h) (i)


(f)
Urinary 2-oxoglutarate Urinary taurine Urinary creatinine
Urinary citrate O
O
O OH O O NH
O O
S
HO OH HO NH2
HO OH N NH
O O
OH
CH3

Younger Aged Younger Aged Younger Aged Younger Aged


rats rats rats rats rats rats rats rats

(k) Urinary
(j) Urinary betaine trim ethylamine-N-
CH3 O oxide O-
+
N - +
H3C O N
CH3 H3C CH3
CH3

Younger Aged Younger Aged


rats rats rats rats

Fig. 3. Quantitative variations of different metabolites in different biological matrices


(serum, plasma, urine) in relation to different physiological factors (gender, age, sexual
maturity degree, body weight).

Differences in the concentrations of stool short-chain fatty acids (SCFA) between the lean,
overweight and obese human subjects have been shown to be important (Schwiertz et al.,
2010): the mean of total SCFA concentration in fecal samples of obese volunteers was by
136 Metabolomics

more than 20% higher in total than of lean volunteers. The highest increase was seen for
propionate (Fig. 3e) with 41%.
The age is known to result in numerous physiological changes that are reflected by physical
and metabolic changes:
Previous studies have shown that ageing rats decrease their excretion of citrate and 2-
oxoglutarate versus increase in their taurine and creatinine output (Fig.3f-i) (Bell et al.,
1991). The increase in creatinine with age (up to 6 months) may be associated with the
increase output from the muscle of larger rats as well as an age-related increase in the
glomerular filtration rate (the rat kidney becomes fully functional at 3 months).
Moreover, in young rats (1 month or less), the urinary excretion of trimethylglycine (betaine)
and trimethylamine-N-oxide was higher than in older rats (Fig. 3j, k). The increased level of
betaine and trimethylamine-N-oxide was higher than in older rats. The increased level of
betaine in the urine of young rats may be due to high choline levels (Bell et al., 1991). Betaine
(or N-trimethylglycine) results from oxidation of choline; it represents a reservoir of methyl
groups and plays the role of methyl donor in the synthesis of methionine in mammals.
In plant world, secondary metabolites showed significant variations in relation to species,
age and maturity level: for instance, among two birch species, Silver Birch (Betula pendula)
does not emit sesquiterpenes (SQT), while Downy Birch (B. pubescens) does (Hakola et al.,
2001). Moreover, older trees B. pubescens emitted greater quantities and higher proportions
of SQT than younger ones. In the common snapdragon, Antirrhinum majus, the emission of
methyl benzoate (MeBA) is increased in pollination period (Dudareva et al., 2000); this may
serve as guide for bees to find their way inside the flower. After pollination, emission of
MeBA decreases dramatically.
SCFA are produced by the intestinal microbiota which represent a large part of bacteria
belonging to the phyla of Firmicus, Bacteroides, Actinobacteria, Proeobacteria and
Verrumicrobia (Zoetendal et al., 2008). Some phyla were characterized by high level
production of some metabolites: for instance, Bacteroides phylum produces high levels of
acetate and propionate, whereas several members of the Firmicus phylum produce high
amount of butyrates (Maslowski et al., 2009).

2.3 Metabotypes based on ratios between concentration levels


Metabolic ratios between concentration levels of structurally close metabolites have been
used to characterize different biological states in human populations:
Menstruation affects the N-oxidation of trimethylamine resulting in a fall in the ratio of
trimethylamine-N-oxide on trimethylamine in the urine (Fig. 4a) (Zhang et al., 1996).
A metabolomic study on different body weight human subjects showed that leaner people
had higher ratios of acetate to butyrate and propionate (Fig. 4b) (Schwiertz et al., 2010,
Duncan et al., 2007).
It has been shown that in a Caucasian population, the urinary metabolic ratio of 6 β-
hydroxycortisol to cortisol was significantly increased in females compared to males (Fig.
4c) (Lutz et al., 2010). This ratio is used as an endogenous marker for CYP3A activity.
Metabotype Concept: Flexibility, Usefulness and Meaning in Different Biological Populations 137

Metabolites' pair Biological matrix Physiologic state ratio

trim ethylamine-N-
oxide Out of menstruation
O-
N
+ Increase
H3C CH3
(a) CH3
Urinary ratio
N
H3C CH3
CH3 Decrease
trim ethylamine
Menstruation

Metabolites' pairs Biological matrix Physiologic state ratio

Acetate Acetate Lean subjects


O O

- -
H3C O H3C O
(b) Fecal ratio
O O
H3C CH 2 C
- -
O H3C O
Propionate Butyrate
Obese subjects

CH 2OH
6 --hydroxycortisol
C O
CH3
HO OH

CH3 Female Caucasians

(c) O Increase
OH
Urinary ratio
CH 2OH
Cortisol
C O
CH3
HO OH
Decrease
CH3
Male Caucasians

Fig. 4. Metabolic ratios between structurally close metabolites providing biochemical


discriminations between different biological states in human populations.
138 Metabolomics

2.4 Metabotypes based on profiling


In animals, gender, species or races have been characterized by specific profiles of organic acid
derivatives (amine, tricarboxylic) which are produced at relatively high or low levels:
Metabolic profiling has been applied to characterize genders in rats: elevated levels of bile
acid metabolites in urine of female rats reflected increased rate of cholesterol and bile acid
synthesis compared to males (Stanley, 2002).
Metabolomic differences between rats and mice have also been highlighted by NMR: 1H-NMR
spectra of urines from B6C3F1 mice and Sprague-Dawley (SD) rats revealed consistently
higher levels of formate, creatinine, hippurate, dimethylglycine, dimethylamine, fumarate, 2-
oxoglutarate and citrate versus lower levels of taurine and betaine (i.e. N-trimethylglycine) in
the rats compared to mice (Bollard et al., 2005).
1H-NMR spectra of urine samples showed that the genetic strain Alpk:ApfCD mice had
relatively higher levels of 2-oxoglutarate, citrate, trimethylamine-N-oxide and
guanidinoacetic acid, whilst C57BL107 mice had higher levels of taurine, creatinine,
dimethylamine and trimethylamine (Fig. 5) (Gavaghan et al., 1996).

O OH
Metabolomic profile of
O O Alpk:ApfCD mousse strain
HO OH O-
OH
O
O O +
N HN NH
HO OH
H3C CH3 OH
CH3
O NH2
O
NH
O N
H3C CH3
N NH H3C CH3
S CH3
HO NH2
CH3 N
O
H
Creatinine

trim ethylamine
2-oxoglutarate
Citrate

dim ethylamine
Trim ethylamine-N-

Guanidinoacetic

Taurine
acid
oxide

Metabolomic profile of
C57BL107 mousse strain
Creatinine

trim ethylamine
2-oxoglutarate
Citrate

Trim ethylamine-N-

Guanidinoacetic

Taurine

dim ethylamine
acid
oxide

Fig. 5. Metabolomic profiles representing two genetic strains of mice on the basis of relative
levels of several metabolites. Bar heights are indicative of relatively higher or lower
concentrations depending on mousse strains (Gavaghan et al., 1996).
Metabotype Concept: Flexibility, Usefulness and Meaning in Different Biological Populations 139

Metabolic phenotypes analysis has been applied to characterize particular laboratory animal
varieties including "germfree" (GF) specimens. GF is the highest quality level of laboratory
animals in which there are no any detectable microorganisms in contrast to those commonly
known as “SPF”, which is merely free of specific pathogens. Germfree animals are especially
useful in the researches concerning genetic engineering, cancer, normal intestine flora,
immunology and nutrition.
Aqueous extract profiles of gut tissues from GF mice were markedly different from those of
conventional mice (Claus et al., 2008) (Fig. 6):
i. The metabolite profile of the duodenum from GF mice was mainly characterized by
higher levels of tauro-conjugated bile acids (TCBAs) and alanine versus lower levels of
glycerophosphocoline (GPC) (Fig. 6a) when compared with conventional mice.
ii. The jejunal tissue of GF group had higher levels of creatine and TCBAs versus lower
levels of tyrosine (Fig. 6b).
iii. The ileum of GF mice was characterized by a higher level of TBCAs and lower levels of
glutamate, fumarate, lacate, phophocholine and alanine when compared with the ileum
from conventional mice (Fig. 6c).
iv. The metabolic profile of the colon from GF mice revealed a high level in a complex
carbohydrate identified as raffinose, and lower levels of lactate, creatine, 5-aminovalerate,
propionate, glutamine, myo-inositol, scyllo-inositol, GPC, phosphocholine, choline,
formate, uracile and fumarate (Fig. 6d) (Monero and Arus, 1996).
Metabolic profiles of ileum and particularly colon in GF mice were markedly more affected than
those of duodenum and jejunum. This reflects the higher microbial loads found in ileum and
colon (Dunne, 2001). The lower levels of choline and its phosphorylated derivatives, GPC and
phosphocholine (Fig. 6a, c-d) were reported to be likely due to the disturbance of the membrane
of colonocytes in GF mice (Claus et al., 2008). Also, the accumulation of the trisaccharide,
raffinose, can be a possible consequence of this disruption. In GF animals, raffinose seems to be
able to cross the epithelial membrane and accumulates in colonocytes where it induces a rise in
osmotic pressure. This phenomenon provokes a well-known signaling cascade that leads to the
release of the mobile osmolytes: GPC, myo-inositol and scyllo-inositol.
Beyond static analysis, kinetic metabolic profiling is applied in chronobiology and
pharmacokinetics in relation to intrinsic or extrinsic factors (e.g diurnal variations):
In SD rat, 1H-NMR profiles of urinary samples collected during the day showed lower levels
of hippurate, taurine, and creatinine together with elevated levels of glucose, succinate,
dimethylglycine, glycine, creatine and betaine compared with urine collected during the
night (Bollard et al., 2001).
Male rats secrete growth hormone in an "on-off" episodic rhythm between which there
are periods when there are no detectable levels of the hormone. Growth hormone
secretion in the female rat is "continuous" since hormone levels are always present
(Czerniak, 2001).
In women subjects, plasmatic cortisol stimulated by synacthen (synthetic ACTH) showed
obesity level-dependent kinetic profiles (Fig. 7) (Semmar et al., 2005a): the secretion and
elimination of cortisol were more rapid and higher in the most obese followed by
intermediate obese then non-obese subjects.
140 Metabolomics

O O O
H3C
O O O
S (b)
(a) HO
CH3
N OH

HO
H3C
N
S
OH Duodenum CH3
H
CH3 O Jejunum
CH3 H O
CH3 CH3 H2N N
CH3 H3C OH
OH
NH
NH2 HO OH
HO OH O
O
CH3
H3C + OH
N P
H3C O O OH NH2
- HO
O OH

Tauroconjugated Alanine Glycero- TBCAs Creatine Tyrosine


bile acids phosphocholine

O O O
H3C
S
HO
CH3
N
H
OH (c) Ileum
CH3
CH3 O O
H3C CH3
H3C OH
+
OH N P O O
H3C O
HO OH O O OH HO
NH2 H3C
OH
OH
HO OH
O
NH2 OH

TBCAs Alanine Glutamate Phosphocholine Fumarate Lactate

OH OH

O
HO
HO
O OH
O
OH

O
(d) Colon
HO HO OH
HO
HO
O
O
OH H3C CH3 O
OH OH
HO +
N P HO O O HO OH
H3C O OH
CH3 O OH H3C OH
O OH O NH OH
H2N N O O H3C CH3
OH
OH H3C CH 2 C HO OH
+ H2N O- OH N O HO
N - OH
NH
H2N OH
H3C OH O H
NH2 O

Raffinose Creatine Glutamine Phosphocholine Choline 5-aminovalerate Fumarate Lactate Propionate Uracile M yo-inositol Scyllo-inositol

Fig. 6. Metabolomic profiling of different gut tissues of germ free (GF) mice (Monero and
Arus, 1996). Bar heights are indicative of relatively higher or lower concentrations in GF
compared with conventional mice (details are given in text).
Metabotype Concept: Flexibility, Usefulness and Meaning in Different Biological Populations 141

250

(3)
200
concentrations ng/mL)

(2)
Plasma cortisol

150
(1)

100

50
0 10 20 30 40 50 60

Time (min)
Fig. 7. Kinetic profiles of plasma cortisol concentrations of three body weight levels women
populations, highlighting compensatory process between secretion and elimination in
relation to obesity levels. (1), (2), (3) corresponds to non-obese, intermediate obese and
extreme obese populations, respectively (Semmar et al., 2005).

3. Dietary metabotypes
3.1 Metabotypes based on occurrence of metabolites
Fruits and legumes can be generally characterized by occurrences of specific or abundant
secondary metabolites belonging to flavonoids and terpenoids. For instance, in flavonoid
class, flavonols, flavones, flavanones, isoflavones, flavanols and anthocyanins are widely
present in onions, parsley, citrus fruits, leguminous plants, green tea and blackberry,
respectively (Fig. 8) (Majewska et al., 2011; Holden et al., 2005; Kaufman et al., 1997).
Among the flavonols, quercetin is widely present in the plant world. It occurs as different
glycosidic forms with quercetin-3-rhamnoglucoside (or rutin) being one of the most
widespread forms (Fig. 9d). The different forms of quercetin glycosides have been found to
be good markers in food quality control: in onions, quercetin is bound to one or two
glucoses to give quercetin-4’-glucoside and quercetin-3,4’-glucoside (Fig. 9c); apples and
berries, however, have been characterized by the occurrence of quercetin-3-galactoside and
quercetin-3- arabinoside, respectively (Fig. 9b, a) (Kühnau, 1976; Zheng et al., 2003).
142 Metabolomics

Flavanones Oranges Flavones Parsley

O Lemons O Celery

Grapefruits Chicory

Tomatoes Letuce
O O

Green tea Flavonols Onion


Flavanols Apricot Broccoli
O
O Broad bean Chicory
OH
Chocolate Letuce
OH O

Isoflavones Soya bean


Anthocyanines Blackberry
O Broad bean
Blueberry
+
O Clover
Strawberry
Peanut
O-glycosyl Plum O

Fig. 8. Metabolic characterization of different dietary plants on the basis of high occurrences
of produced flavonoid classes in their tissues.
OH

4' OH

HO 7 O
Quercetin
3
5 OH
OH O

(a) (b) (c) (d)

Blueberries Apples Onions Abundant in plant world

OH
OH OH OH
HO
OH OH CH3
O OH
HO O OH O
O OH
OH
HO O HO OH OH
OH HO O OH
O
O O CH 2OH O
HO O
OH O OH O OH O OH
OH Quercetin-4'- O
OH O OH
OH OH glucoside OH O OH

Quercetin-3-arabinoside Quercetin-3-galactoside OH O
Quercetin-3-(rhamnosyl-
HO 6→1-glucoside)
OH O
OH
O OH
OH (Rutin)
HO O HO Quercetin-
O
OH 3,4'-glucoside
O OH
OH
OH O

Fig. 9. Metabolic characterization of some dietary plants on the basis of abundant quercetin
glycosides in their tissues.

Flavanones are flavonoids particularly abundant in citrus and vary qualitatively in relation
to fruit types (Mouly et al., 1998; Kawaii et al., 1999; Gattuso et al., 2007): The lemon (Citrus
limon) can be distinguished by production of eriocitrin and hesperidin (Fig. 10 a), whereas in
grapefruits, naringin predominates in presence of narirutin (Fig. 10c). In oranges (Citrus
sinensis) and mandarins (Citrus reticulata), hesperidin is the major flavanone in presence of
narirutin (Fig. 10b).
Metabotype Concept: Flexibility, Usefulness and Meaning in Different Biological Populations 143
HO HO
OH OCH3
OH
HO O HO O
HO O
Eriodictyol Hesperetin Naringenin

HO O
HO O
HO O

HO HO
HO
HO OH HO OH
O O OH
H3C OH H3C OH HO O
H3C OH
OCH 3 HO
O OH O
O OH
O O O
HO
O O O O O O
HO HO O HO
HO O O O
HO HO OH
OH
HO OH
H3C O OH O
HO OH
OH O OH O
OH O HO
Eriocitrine Hesperidin Narirutin Naringin

Lemons Oranges, Mandarins Grapefruits

(a) (b) (c)

Fig. 10. Metabolomic distinction between different Citrus species based on major flavanone
glycosides in fruit juices.

3.2 Metabotypes based on concentration levels


High amounts or concentrations of secondary metabolites have been used to control dietary
plant varieties as well as to diagnose animal or human diets:
Onions are rich source of quercetin-4’-O-glucoside and quercetin-3,4’-O-diglucoside. Mullen
et al. (2006) reported that 270 g of lightly fried onions contains 275 µmol of flavonol
glucosides with the main constituents being 143 µmol of the 4’-O-glucoside and 107 µmol of
the 3,4’-O-diglucoside (Fig. 11).

Input Ouput

143 µmol 107

others
275 µmol flavonol µmol
270 g onion
glucosides Q-(4'-O- Q-3,4'-O-
glucoside) glucoside

Source ? ? ? Content
Prediction of dietary source (Diet control)

Fig. 11. Abundance of flavonol glucosides in onion particularly dominated by 4’-O-


glucoside and 3,4’-glucoside of quercetin (Q) (Mullen et al., 2006).

In human subjects, plasma quercetin was found to be a good marker of dietary intake
because its concentrations increase with increasing ingested dose (Radtke et al., 2002). In
a strictly controlled dietary intervention study, 77 health human subjects consumed
either 170 or 850 g of fruits, vegetable and berries daily. Quercetin intake was calculated
to be 3 to 24 mg/d on the respective diets. The mean  SD of plasma quercetin
concentration was 78  56 nmol/L during the habitual diet; it decreased to 70%
during the low-vegetable diet and increased to 170% during the high-vegetable diet
(Freese et al., 2002) (Fig. 12).
144 Metabolomics

Diet mass Diet content Plasma content

170 g: 3 mg 55 nmol/L
(fruits, quercetin plasma
vegetable, intake quercetin
berries)

850 g: 133 nmol/L plasma


24 mg
(fruits, vegetable, berries) quercetin
quercetin intake

Quantitative diet control from plasma content

Fig. 12. Increase in plasma quercetin concentration (nmol/L) in relation to diet mass (g) and
its total quercetin content (mg) in human population (Freese et al., 2002).

Ingestion of 200 mL of coffee has been reported to increase the plasma conjugated caffeic
acid level in human subjects (Nardini et al., 2002).
Following consumption of both green and black tea, human urinary samples showed
significant increases in level of hippuric acid and 4-hydroxyphenylacetic acid (Mulder et al.,
2005).
Excretion of creatinine in the urine of rats, per unit of skeletal muscle mass, was found to be
promoted by food deprivation (Rikimaru et al., 1989).

3.3 Metabotypes based on ratios between concentration levels


Legumes are known to be important dietary sources of isoflavones (Liggins et al., 2000).
Women subjects having consumed isoflavones from soymilk powder were classified into
two excretion levels according to the relative contents of daidzein and genistein recovered in
feces and urine (Fig. 13) (Xu et al., 1995): in strong isoflavone excreters, the percentages of
daidzein and genistein recovered in feces were 10 and 20 times greater than those weak
excreters. Isoflavones recovered in urine for 48h revealed excretions of daidzein and
genistein which were 2 and 3.5 times in high excreters than low excreters.
In analogous study, human subjects consumed a single dose of strawberries (250 g),
raspberries (225 g) and walnuts (35 g), all of which contain ellagitannins (hydrolysable
tannins). Intakes resulted in urinary excretion of a derivative, urolithin B-3-O-glucuronide,
in quantities equivalent to 2.8% (strawberries), 3.4% (rasperries) and 16.6% (walnuts)
regarding the ingested ellagitannins (Fig. 14) (Cerda et al., 2005).
Metabotype Concept: Flexibility, Usefulness and Meaning in Different Biological Populations 145

(a) (b)
HO O
Daidzein analyzed in feces Daidzein analyzed in urine

O
OH
Daidzein

Excretion levels Excretion levels

Genistein analyzed in feces Genistein analyzed in urine

HO O

OH O
OH

Genistein

Excretion levels Excretion levels

Fig. 13. Metabolomic classification of human subjects into two excretion levels of isoflavones
(daizein and genistein) according to percentages of faecal and urinary isoflavones
compared to diet dose. Low and high excreters had low and high excretion percentages of
isoflavones, respectively.
146 Metabolomics

Intake

Strawberries (250 g)

Rasperries (225 g) Microflora (MF) metabolism Urinary Excretion

Walnuts (35 g)
OH
OH OH OH OH Urine
HO HO HO
190 mg
O
422 mg O O O O
191 mg O
Hydrolysis
O MF O MF O MF Glucuronidation
Ellagitannins O O

(MF)
OH OH
OH
O HO
OH OH OH O OH
OH
COOH

Ellagic acid Urolithin D Urolithin C Urolithin B Urolithin B-3-O-


glucuronide

16.6 %

3.4 %

2.8 %

% ingested ellagitannins

Fig. 14. Characterization of three tannin-rich diets by the excretion percentages of a urinary
metabolite (urolithin B-3-O-glucuronide) referred to the ingested dose of ellagitannins
(Cerda et al., 2005).

3.4 Metabotypes based on profiling


Flavanone enantiomers profiles have been analysed in different fruit juices and their
concentrations have been found to be efficient biomarkers of the plant dietary source (Fig.
15) (Yanez et al. 2008):
- Orange juices contained the highest concentrations of (2R)- and (2S)-hesperidin;
- Conventional and organic grapefruit juices contained the highest concentrations of
R(+)- and S(-)-hesperetin, and (2R)- and (2S)-naringin.
- Conventional and organic tomato juices showed the highest levels of R(+)- and S(-)-
naringenin. Also, the chemical profiles of tomato juices showed the co-occurrences of
the eight enantiomers. Organic juice can be distinguished by relatively higher levels of
naringin enantiomers.
Metabotype Concept: Flexibility, Usefulness and Meaning in Different Biological Populations 147

O O
R-(+)-enantiomer S-()-enantiomer

O O

Conventional Organic
orange juice orange juice

Conventional Organic
grapefruit juice grapefruit juice

Conventional Organic
tomato juice tomato juice

Fig. 15. Metabolomic characterization of different fruit juices by their chiral flavanone
profiles.
148 Metabolomics

Human subjects ingesting three times two cups of coffee at 4-h intervals had urinary profiles
containing ferulic, isoferulic, dihydroferulic, 3-methoxy-4-hydroxybenzoic, hippuric and 3-
hydroxyhippuric acids (Rechner et al., 2001).
After ingestion of 270 g of lightly fried onion by human subjects, plasma and urine samples
collected over 24h showed very different metabolic profiles of concentrations (Fig. 16)
(Mullen et al., 2006):

(a) plasma (b) urine


OH
OH
Quercetin-3'-O-
-
OSO3 Isorhamnetin-4'-O- OCH3 Quercetin-3'- HO
O OH
HO
sulfate OH glucuronide O
O OH glucuronide O COOH
COOH
OH
HO O HO O
HO O

OH OH
OH
OH O OH O
OH O

Fig. 16. Metabolic profiles of conjugated quercetin metabolites in plasma (a) and urine (b)
following ingestion of lightly fried onion (270 g) by health human subjects (Mullen et al.,
2006).

The main plasma metabolite, quercetin-3’-O-sulfate, was excreted only in trace quantities in
urine while isorhamnetin-O-glucuronide and quercetin-O-diglucuronide that were minor
components in plasma were major urinary metabolites (Fig. 16). Several other metabolites,
including quercetin-3’-O-glucuronide and isorhamnetin-4’-O-glucuronide, which were
present in trace quantities or absent from plasma were excreted in urine in substantial
amounts.
In two separated human studies, and following the consumption of 200 g of strawberries
(Mullen et al., 2008) or 200 g of blackberries (Felgines et al., 2005), the urinary contents were
Metabotype Concept: Flexibility, Usefulness and Meaning in Different Biological Populations 149

characterized by pelargonidin and cyanidin metabolites profiles, respectively: following the


consumption of strawberries, urine samples were characterized by the predominance of
pelargonidin-3-O-glucose in presence of pelargonidin-O-glucuronides with small quantities
of pelargonidin aglycone and a pelargonidin-O-sulfate. Following the consumption of
blackberries, the urine had a cyanidin-based profile containing unmetabolized cyanidin-3-
O-glucoside, a cyanidin-O-glucuronide and a 3’-O-methyl-cyanidin-O-glucuronide.
In obese human population, low and medium carbohydrate diets reduced the total SCFA
concentrations in fecal matrix, compared with a maintenance (high carbohydrate) diet
(Duncan et al., 2007). This decrease concerned also acetate, propionate, and valerate
concentrations analysed in saddles (Fig. 17). Low and medium carbohydrate diets can be
distinguished by lower butyrate concentrations; such a reduction is more marked under low
diet than intermediate one.

(a) (b) (c)

Fig. 17. Variations in concentrations of short chain fatty acids (SCFA) and lactate in stool
samples of obese humans in relation to three carbohydrate levels diets (Ducan et al., 2007).
(a), (b), (c) correspond to high, medium and low carbohydrate diet, respectively.

A previous study showed that SD rats deprived of water for 48h had elevated levels of
creatinine and depleted levels of taurine, hippurate, 2-oxoglutarate, succinate and citrate
(Clausing and Gottschalk, 1989). Water deprivation has a direct effect on osmoregulation
implying variations in osmoregulators’ levels as taurine.

4. Clinical metabotypes
Pathologies are known to induce changes in concentrations, regulation ratios and overall
profiles of different metabolites that could be used to diagnose or characterize different
diseases. Some examples will be given to illustrate the interest of different metabolomic
criteria in clinical cases.

4.1 Metabotypes based on occurrences of metabolites


Occurrences of particular metabolites in biological matrices represent a strong metabolomic
parameter to identify intoxication or doping sources:
150 Metabolomics

Among endogenous metabolites, a particular attention has been paid to glutathione


conjugates as potential markers of exposure (Van Weli et al., 1992). This is because
glutathione (GS-H) detoxifies reactive chemicals (R-X) to which biological systems are
exposed. The result of this conjugation (GSR) is the excretion of a variety of sulphured
metabolites.
Concerning xenobiotics, chemicals that are reactive or are metabolized to intermediates
reacting with DNA are of particular concern in relation to genotoxicity, and therefore,
possible carcinogenicity:
In styrene industry, exposure to reactive alkylating agents can be diagnosed by analyzing
DNA adducts, such as styrene oxide-O-6-guanine detected in white blood cells of exposed
workers (Hemminki and Vodicka., 1995). Exposure to cyanide can be identified by rapid
analysis of this toxin in blood of exposed individuals. However, free cyanide disappears
rapidly from blood suggesting that a biological sample should be collected quickly, and
analysis should be performed as soon as possible. If analysis of cyanide cannot be
performed quickly, then cyanide exposure can be identified from the detection of three
major markers of cyanide in the blood or urine: cyanide ion (CN-), thiocyanate (SCN-), and
2-aminothiazoline-4-carboxylic acid (ATCA) or its tautomer 2-iminothiazolidine-4-
carboxylic acid (ITCA) (Logue et al., 2005).
Adducts such as N-(2-hydroxyethyl)valine have also been detected in haemoglobin from
hospital workers exposed to ethylene oxide (Van Welie et al., 1992).
In clinical field, DNA adducts have also been detected in the white blood cells and urine of
patients treated with anti-cancer drug such as N-methyl-N-nitrosourea (Prevost et al., 1996).
Also, the N-7-guanyl aflatoxin B1 adduct can be detected in urine, and used as a biomarker
of exposure to the carcinogen aflatoxin B1, which may be present in the diet (Groopman et
al., 1994).
Oxidative damage to DNA can be detected by urinary 8-hydroxy-2’-deoxyguanosine (Van
Welie et al., 1992). Also, urinary 8-hydroxy-2’-deoxyguanosine has been proposed as a
biomarker of oxidative stress in humans (Bianchini et al., 1996).
In sport, doping controls are generally carried out on the basis of two complementary tests:
(i) urine analysis and blood analysis provide short-term information on drug use by an
individual; (ii) however, long term histories are accessible through hair analysis, because
drug appears to be incorporated into the hair. For instance, in complement of testosterone
determination, the identification of unique testosterone esters in hair enables an
unambiguous charge for doping because the esters are certainly exogenous substances
(Gaillard et al., 1999).
For drugs of abuse like cocaine and opiates, the threshold dose for detecting cocaine in hair
appears to be approximately 25-35 mg cocaine, administered intravenously (Henderson, et
al.; Kintz et al., 1999). Once incorporated into hair, a single dose of cocaine can be detected
for 2 to 6 months. Codeine was detected in hair for 8 weeks after a single oral dose of 60 mg
(Thieme et al., 2000).
Following intranasal absorption of 1.5 mg/kg BW of cocaine hydrochloride by humans,
urinary excretion of unchanged cocaine was detected for only 8h (maximal excretion within
Metabotype Concept: Flexibility, Usefulness and Meaning in Different Biological Populations 151

2 h) (Hamilton et al., 1977). However, benzoylecgonine (the major hepatic metabolite of


cocaine) was generally detected in urine for 48 to 72 h (maximal excretion 4 to 8 h following
cocaine administration).

4.2 Metabotypes based on concentration levels


The NMR spectrum of a biofluid can be conveniently thought of as a series of ‘biomarker
windows’, which are spectral regions that contain signals from metabolites associated with
specific targets for toxicity or disease (Fig. 18). For instance, metabolic response of the
multimammate mousse (Mastomys natalensis) to 2-bromoethanamine (C2H6BrN) and
propyleneimine (C3H7N) treatment was the induction of taurinuria (Fig. 18a). Taurine is
amino acid known to protect renal medullary cells from osmotic stress and therefore may
accumulate in the inner medulla of the Mastomys, protecting it from nephrotoxicity (Holmes
et al. 1997).
Animal species Toxins Target organs Urinary metabotypes

2-bromoethanamine
O
Taurine
Mastomys natalensis
(a) Kidney disturbance S
(mousse) HO NH2
O

propyleneimine

Creatine TCA
intermediate
(b) SD rat 2-aminoadipate O CH3 O H 2C COO
-

-
O H2N N
Hydrazine Liver O
-
OH HO C COO
-

(steatosis) O NH2 NH
B6C3F1 mousse H 2C COO
-

Carbon
tetrachloride Creatine
O
Taurine CH3 O
(c) Rats Reduced S H2N N
Thioacetamide liver function HO NH2 OH
O
NH

Allyl alcohol

Fig. 18. Variation of urinary concentrations of metabolites, indicating kidney or liver


perturbations in animal species previously exposed to some toxins.

Combining 1H-NMR urine spectra with multivariate statistical analysis, metabolic


responses of SD rats and B6C3F1 mice to hydrazine (NH2NH2) exposure were
investigated (Bollard et al., 2005). Several common metabolic responses to hydrazine
consisted of elevated levels of 2-aminoadipate and creatine versus depletion of the TCA
cycle intermediates in the urine (Fig. 18b). Combined increases in taurine and creatine
concentrations in urine have been associated with reduced liver function after exposure of
rats to carbon tetrachloride (CCl4), thioacetamide (C2H5NS) and allyl alcohol (C3H6O) (Fig.
18c) (Holmes et al., 1998).
In oncology, human prostatic epithelial cells are unique in accumulating zinc that blocks
citrate degradation. In cancer cells, however, the prostate tissue contains low levels of citrate
because it does not accumulate zinc and because most of citrate is used for fatty acid
synthesis. 1H-NMR of prostate tissues confirmed the dramatic decrease in citrate levels in
prostate gland during malignancy (Raina et al., 2009). A recent study showed a lower risk of
152 Metabolomics

developing high-grade prostate cancer for men with low serum cholesterol levels (Platz et
al., 2009). Inversely, high levels of cholesterol in metastasic bone tissue (>70mg/g tissue)
were found to be revelator of prostate cancer, compared to normal bone tissue (50-60mg
cholesterol/g tissue) as well as to bone metastases from other cancers (<70mg/g) (Thysell et
al., 2010).

4.3 Metabotypes based on ratios between concentration levels


Induction of cytochrome P450 isozymes may be used as biomarker of the effect of
exposure of many species to a variety of chemicals, such as organochlorine compounds
and polycyclic hydrocarbons. There are well-established urinary markers for cytochrome
P450 induction, such as increased ratio of 6--hydroxycortisol/17-hydroxycorticosteroids
(6β-OHF/17-OHCS ) (Hugget et al., 1992). For instance, in a recent study, serum
carbamazepine level was inversely associated with the urinary 6β-OHF/17-OHCS ratio
(Konishi et al., 2004).
Elevated urinary ratio of creatine/creatinine has been proposed as marker of testicular
damage (Timbrell et al., 1994).
Important increase of the ratio of lactate level on glucose level can be a biomarker of cancer
cells in biological body: the conversion of glucose to lactate in the presence of oxygen
represents a critic aerobic pathway that allows cancer cells to proliferate rapidly (Kim and
Milner, 2011; Mazurek et al., 2011). Cancer cells metabolize glucose and glutamine more
than normal cells to support the de novo biosynthesis of nucleotides and energy required for
the high rate of cell proliferation.

4.4 Metabotypes based on profiling


Metabolomic profiling has been used to reliably identify different diseases including cancers
and cardiovascular disturbances:
Concerning breast cancer, malignant cells (MDA-MB-435) content showed significant
increase in glutathione (GSH) , m-inositol, creatine and phosphocholine concentrations and
decrease in isoleucine, leucine, valine, and taurine concentrations, compared to normal
mammary epithelial cells (MCF-10A) (Fig. 19) (Yang et al., 2007).
However, free choline and glycerophosphocholine were below the detection level in MDA-
MB-435.
In patient suffering from coronary artery disease or left ventricular dysfunction,
preischemia state was characterized by higher alanine levels versus lower concentrations
of glucose, lactate, free fatty acids, total ketones, 3-hydroxybutyrate, pyruvate, leucine
and glutamate analysed in the coronary sinus compared with arterial sample contents
(Turer et al., 2009).
In patients suffering from non-ST-segment elevation acute coronary syndrome, plasma
samples showed decrease in citric acid, 4-hydroxyproline, aspartic acid and fructose versus
increase in lactate, urea, glucose and valine, compared to control healthy subjects (Vallejo et
al., 2009).
Metabotype Concept: Flexibility, Usefulness and Meaning in Different Biological Populations 153

Fig. 19. Metabolomic profile characterizing breast tumor cells (MDA-MB-435) from normal
cells (MCF-10A) based on ratios between metabolite concentrations MDA-MB-435/MCF-
10A. High metabolic regulations are indicated by ratios >1, and inversely.

5. Metabotypes linked to biodiversity and environment conditions


5.1 Metabotypes based on occurrences of metabolites
In plant world, presence-absence of some secondary metabolites have high chemotaxonomic
values. For instance, in monocotyledons, some families (Poaceae and Cyperaceae) are
characterized by C-glycosyl flavonoids, i.e. flavonoids in which aglycone and saccharidic
moiety have a C-C link, in addition to the C-O link which is abundant in the plant world
(Fig.20a) (Semmar, 2010 ).
154 Metabolomics

(a) (Poaceae) (b) (Liliaceae)

O-C link HO
OH
HO OH
HO O
OH
HO
Sugar(s)-O O
OH
O O
OH

HO O
O-Sugar(s)
HO
HO O
OH HO O
flavonol-3,7-O-diglycoside
C-C link

C-glycosyl-flavones (d) Family level


Lamiaceae

(c) (Fabaceae) HO O
OH
OH

O
H3CO
HO
OH O
O
R=H R H
O OH O
H3C HO
O
HO
O HO O Lamioideae Subfamily level Nepetoideae
O OH
HO
O OH
HO OH OH O COOH
No R.A. HO
HO OH O OH

HO
Rosmarinic acid (R.A.)
flavonol-3-O-tetraglycoside

Tribe level
(Asteraceae)
(e) Saturejeae Other tribes
OH
OH H3CO O
H3CO
HO O H3CO O No HMF
HO
OCH 3 O
HO OH HO
HO O
OH O OCH 3

Quercetagetin Aurones 5,6-diOH, 7,8-diOMe flavone (HMF)

Fig. 20. Phytochemical characterizations of different plant taxons on the basis of specific or
abundant phenolic compounds in their tissues.

Apart the link type between aglycone and sugar, glycosylation degree provides good
chemotaxonomical criterion to a general characterization of plant families. For instance, leaf-
tissues of Liliaceae species (monocotyledons) were characterized by the presence of di- and
tri-O-glycosides of flavonoids, and a rare occurrence of monoglycosides (Williams, 1975).
Flavonol 3, 7-diglycosides seem to be common constituents of the Liliaceae (Fig.20b)
(Budzianowski, 1991; Williams, 1975). The family Fabaceae (dicotyledons) has been shown
to be productive of multiglycosylated flavonols (Fig.20c) (Semmar, 2010).
Within the Lamiaceae family (dicotyledons), the presence-absence of rosmarinic acid (Fig.
20d) has been shown to be an excellent chemotaxonomic marker because of its presence in
the subfamily Nepetoideae and absence in the subfamily Lamioideae (Janicsák et al., 1999;
Harborne, 1966b). Members (tribes and genera) of these two subfamilies have been
phytochemically characterized by the presence-absence of hydroxyl and methyl groups
substituted on the A-ring of flavone aglycones. For instance, the presence of 5,7-dihydroxy-
6-methoxyflavones with a substituted B-ring is characteristic of the subfamily Nepetoideae
(Fig. 20d), particularly of Salvia, Rosmarinus and Ocimum species (Tomás-Barberán and
Wollenweber, 1990); in Lamioideae, a 5,7-dihydroxy 6-methyl ether flavone has been found
Metabotype Concept: Flexibility, Usefulness and Meaning in Different Biological Populations 155

in the genus Scutellaria but with unsubstituted B-ring (Fig. 20d). Moreover, in the subfamily
Nepetoideae, the genera Thymus, Satureja, Micromeria, Acinos, Calamintha, Origanum and
Mentha were characterized by the production of the 5,6-dihydroxy-,7,8-dimethoxyflavone
(Tomás-Barberán and Wollenberg, 1990); all these genera belong to the tribe Saturejeae (Fig.
20d).
Among the dictotyledons, the family Asteraceae has been characterized by the production of
aurones and quercetagetin which is a flavonol almost entirely found in this family (Fig. 20e)
(Iwashina, 2000).
In Tulipa (Liliaceae), the flower colors are fundamentally defined by the anthocyanidin type:
orange and flesh pink colors are linked to pelargonidin, black-red and red-orange are due to
cyanidin, and black-blue-violet-purple are governed by delphinidin (Fig. 21) (Shibata and
Ishikura, 1960; Torskangerpoll et al., 2005).
Moreover in tulips, the shade of flower tepals was showed to be dependent on some
chemical substitutions of anthocyanins: substitutions of anthocyanins by aromatic acyl
groups have been reported to be responsible for bluing effect (Torskangerpoll et al., 2005).
Also, combinations of cyanidin and pelargonidin with carotenoids generally induced
attractive red and orange colours (van Eijk et al., 1987).

5.2 Metabotypes based on concentration levels


In plant world, accumulation of anthocyanins has been shown to be a good marker of cold
stress: low temperatures have been shown to induce anthocyanin synthesis in many plant
species, e.g. in leaves of Arabidopsis thaliana (Leyva et al., 1995), Cotinus coggygria (Oren-
Shamir and Levi-Nissim, 1997), Pinus banksiana (Krol et al., 1995), etc. .

Pelargonidin Cyanidin Delphinidin


HO
OH OH HO
OH
HO O+ HO O+
HO O+
OH
OH OH
OH
HO HO
HO

Orange, flesh pink Black, violet, purple


Black-red, red-purple
dark red-orange, red-
orange

Fig. 21. Variation of tulip flower colours in relation to occurrence and abundance of
anthocyanidin type.
156 Metabolomics

Apart anthocyanins, sesquiterpenes (SQT) were found to be reliable indicator of thermic


stress in emitting plant species: for instance, in young orange tree, emission of -
caryophyllene (-Car) increased 5-6 fold for air temperature increase of 10°C (Hansen and
Seufert, 1999). Temperature experiment performed on young corn plants showed that the
proportion of -Car (as a percentage of total emitted biogenic volatile organic compounds)
was maximal at 37°C (Gouinguené and Turlings, 2002).

SQT levels have been also found to be biomarkers of diurnal and seasonal rhythmicity: -
Car emissions from various Citrus varieties were found to increase during the morning
with generally a concentration peak around noon (Ciccioli et al., 1999). In potatos, emitted
SQT increased steadily throughout the day and peaked in the afternoon (Agelopoulos et
al., 2000). In Finnish scots pine, -Car emissions exhibited significantly seasonal
variability, with maximum emissions observed during summer months (Tarvainen et al.,
2005).
Volatile terpenes (isoprenes, monoterpenes and sesquiterpenes) were found to be good
markers of water stress in some plants. In Pinus halepensis (Alepo pine), water stress induced
monoterpenes emissions by the leaves (Ormeño et al., 2007a). In young orange tree, severe
drought reduced -Car emissions to 6% of pre-drought levels, but emissions were
unaffected by mild drought conditions (Hansen and Seufert, 1999).
Different works concluded relationships between metabolic variability in plants and soil
composition: higher concentrations of aluminium in soil resulted in increase in exuded
phenolic compounds by the roots of maize (Kidd et al., 2001). Aluminium resistant variety
of maize exuded 15-fold higher level of flavonoids when pre-treated with silicon than
when no such pre-treatment was applied. In scots pine (Pinus sylvestris L.), tree exposed to
nickel had higher concentrations of condensed tannins compared with control (Roitto et
al., 2005). Calcareous soils stimulated emissions of -humulene from Aleppo pine,
whereas siliceous soils favored -humulene and -bourbonene from Rock Rose (Ormeño
et al., 2007b).
In plant world, emissions of some volatile compounds were found to be positively
correlated to biotic disturbance such as parasite or herbivory: in Black Sage, SQT
emissions increase significantly under infection with aphids (Arey et al., 1995). In corn
seedlings, SQT emissions increase as a response to caterpillar feeding, and it has been
demonstrated that such emission attracted wasps which parasitize caterpillars (Turlings et
al., 1995).

5.3 Metabotypes based on ratios between concentration levels


Several plant species have been biogenically characterized by the ratios of individual
sesquiterpenes (SQT) relative to the overall emitted (volatile) SQT (Duhl et al., 2008). In SQT
emission profiles, -caryophyllene (-Car) was the most frequently reported and abundant;
- and - farnesene as -humulene are also prominent components to observed profiles (Fig.
22). The results concern studies where the plants were not disturbed because, disturbance is
known to affect the variability of emitted SQT blends:
Metabotype Concept: Flexibility, Usefulness and Meaning in Different Biological Populations 157

CH3 CH3
CH3
CH3

CH3
CH2
CH3 H3C
CH2 CH2 H2C
H H
H3C
H3C H3C CH3
CH3 H3C

-Caryophyllene -Humulene -Farnesene Germacrene D


Plant species

% emitted SQT % emitted SQT % emitted SQT % emitted SQT

Fig. 22. Characterization of different plant species by the percentages (%) of some volatile
sesquiterpenes (SQT) (-caryophyllene, -humulene, -farnesene, germacrene D) referred to
the overall SQT emissions.

SQT ratios showed that plant species can be characterized by dominance or relatively high
levels of some SQT : sunflower, hormbeam and citrus species are highly productive of -
caryophyllene (-car) (>90%). The SQT pool of gray pine seems to be dominated by high
relative levels of -farnesene (77%); the corn shows wide inter-individual variation range of
-farnesene going from 0 to 70%. Marsh elder can be characterized by germacrene D
representing 48 to 54% of overall emitted SQT. Trembling aspen seems to emit relatively
more -humulene with high inter-individual variability (3-36%). Apart from -humulene, -
humulene (not presented) is also frequent in nature and was reported to represent 55-57% of
SQT in red and white pines (Duhl et al., 2008).
In tulips (Liliaceae), the flower colors acquire higher variability governed by the relative levels
of mixed anthocyanins (Shibata and Ishikura, 1960): cultivars having "magenta nuances"
showed anthocyanin content in which the relative amounts of cyanidin 3-rutinoside increased
at the expense of delphinidin-3-rutinoside. Garden varieties with blue nuance (black, black-
purple, fade-sky, violet and purple) have relatively high content of delphinidin type in tepals
(i.e. the delphinidin content was more than 50% of the total anthocyanin content). Orange
colored tepals were to a large extent correlated with high relative amounts of the pelargonidin
derivatives at the expense of the two other aglycone types.
Apart from the chemotaxonomic characterization of plants, metabolic ratios were analysed
in relation to different environmental conditions to characterize adaptive responses of
biological species:
158 Metabolomics

By opposition to monoterpene emissions, sesquiterpene emissions were found to be reduced


by water deficit in Pinus halepensis among other typical Mediterranean species (Ormeno et
al., 2007a). On the basis of negative effect on sesquiterpene emissions versus positive effect
on monoterpene emissions, the drought stress can induce a shift in terpene composition
leading to an increase in the ratio monoterpenes/sesquiterpenes in plant.

5.4 Metabotypes based on profiling


Metabolic profiling is used in chemotaxonomy to highlight chemical polymorphisms in
plant species leading to better understand the biochemical origins of biodiversity:
For instance, analysis of flavonoid glycosides in the leaves of Astragalus caprinus (Fabaceae)
highlighted four chemotypes (Chtp) characterized by high relative levels of different
compounds among 14 in all (Fig. 23) (Semmar et al., 2005b): Chtp I was exclusively
characterized by the presence of diglycosides of methylated flavonols (rhamnazine,
rhamnocitrin) (11-14) which are acylated by a methyl-glutaric acid. Chtp II was
characterized by high regulation of a tetraglycoside of quercetin (1). Chtp III was
characterized by high regulations of a tetraglycoside of kaempferol (2) and its acylated
derivatives (acylated by p-coumaric or ferrulic acid) (6-10). Chtp IV was characterized by a
relatively high regulation of a triglycoside of kaempferol (5).
Correlation analyses between the relative levels of the 14 flavonoids showed positive trends
between flavonoids based on a same aglycone and negative trends between flavonoids
having different aglycones (kaempferol, quercetin and methylated aglycones) (Semmar et
al., 2007). This compatibility between statistical correlations and chemical structures helped
to analyse the network of metabolic links between flavonol glycosides (Fig. 23).

FS Dihydro-kaempferol FH

Kaempferol Quercetin
GT
GT
1
Chemotype IV
OH
MT, AT OH
OH
HO O
HO O

O OH O
O Acyl. DiG. Rh. O
O OH O
H3C HO
O 5 H3C HO
HO O
O HO OH HO
O OH
HO O HO O
O OH
HO
HO OH O
HO OH OH
OH
OH

ApT 2
RhT
HO

3 HO O
OH 11-14
O
O HO O
OH
MT
H3 C HO
OH O
H3CO O
Chemotype II
O O
O OH O O
HO O
O HO O H3C HO O OCH3
OH OH O
HO O O OH
O HO HO
HO OH OH O HO O HO CH3 O O
HO OH
HO O OH
O
CH3
OH HO O
4 HO

HO OH OH
OH O O
OH O O OH O
AcT OH
H3C HO
HO OH O
HO
OH O HO
OH O
HO O
6-10 HO O
HO OH
O
OH
Chemotype I
O HO OH
O OH O
H3C HO
O
HO
O O O
O OH
HO CH3
HO OH
O
OH
OH Chemotype I
HO OH
O

Chemotype III

Fig. 23. Four chemotypes of Astragalus caprinus (Fabaceae) based on different metabolic
regulations between flavonol glycosides pathways. FS: Flavonol Synthase; FH: Flavonol
Hydroxylase; Acyl. Dig. Rh.: Acylated Diglycosyl of Rhamnazin or Rhamnocitrin
Metabotype Concept: Flexibility, Usefulness and Meaning in Different Biological Populations 159

Sampling of 404 plants of A. caprinus from North to South of Tunisia highlighted a


significant link between metabotypes’ (chemotypes’) abundances and geographical area
(climatic conditions): Chemotype I rich in less hydrophilic compounds was abundant in the
south (arid). The north (humid) seemed to offer favorable conditions to chemotypes III and
IV (both based on kaempferol derivatives). The center of Tunisia, with intermediate climatic
conditions, allowed a co-evolution of the four chemotypes with a relatively higher
abundance for chemotype II (based on quercetin metabolism).

6. References
Arey, J., Crowley, D. E., Crowley, M., Resketo, M., and Lester, J. (1995). Hydrocarbon
emissions from natural vegetation in California’s South Coast Air Basin. Atmos.
Environ., Vol. 29, pp. 2977–2988, 13522310
Agelopoulos, N. G., Chamberlain, K. & Pickett, J. A. (2000). Factors affecting volatile
emissions of intact Potato plants, Solanum tuberosum: variability of quantities and
stability of ratios. J. Chem. Ecol., Vol. 26, pp. 497–511, 0098-0331.
Barderas, M.G., Laborde, C.M., Posada, M., de la Cuesta, F., Zubiri, I., Vivanco, F. &
Alvarez-Llamas, G. (2011). Metabolomic Profiling for Identification of Novel
Potential Biomarkers in Cardiovascular Diseases. J. Biomed. Biotechnol. 2011, ID
790132, pp. 1-9, 11107243
Bell, J.D., Sadler, P.J., Morris, V.C. & Levander, O.A. (1991). Effect of aging and diet on
proton NMR spectra of rat urine. Magn. Reson. Med., Vol. 17, No. 2, pp. 414–422,
15222594
Bianchini, F., Hall, J., Donato, F. & Cadet, J. (1996). Monitoring urinary excretion of 5-
hydroxymethyluracil for assessment of oxidative DNA damage and repair.
Biomarkers, Vol. 1, pp. 178-184, 1354-750X.
Bishop, J.H. & Green, R. (1980). Effects of pregnancy on glucose handling by rat kidneys. J.
Physiol., Vol. 307, pp. 491–502, 00223751
Bollard, M.E., Holmes, J.C., Lindon, S.C., Mitchell, D., Branstetter, W. & Zhang JK
Nicholson, (2001). Investigations into biochemical changes due to diurnal variation
and estrus cycle in female rats using high resolution 1H NMR spectroscopy of
urine and pattern recognition. Anal. Biochem., Vol. 295, pp. 194–202, 00032697
Bollard, M.E., Keun, H., Ebbels, T., Beckonert, O., Antti, H., Lindon, J.C., Holmes, E. &
Nicholson, J.K. (2005). Comparative metabonomics of differential species toxicity of
hydrazine in the rat and mouse. Toxicol. Appl. Pharmac., Vol. 204, No 2, pp. 135-151,
0041008X
Budzianowski, J. (1991). Six flavonol glucuronides from Tulipa gesneriana. Phytochemistry
30, 1679-1682, 0031-9422.
Cerda, B., Tomas-Barberan, F.A. & Espin, J.C. (2005). Metabolism of antioxidant and
chemopreventive ellagitannins from strawberries, raspberries, walnuts, and oak-
aged wine in humans: identification of biomarkers and individual variability. J.
Agric. Food Chem., Vol. 53, pp. 227–235, 00218561
Ciccioli, P., Brancaleoni, E., Frattoni, M., Di Palo, V., Valentini, R., Tirone, G., Seufert, G.,
Bertin, N., Hansen, U., Csiky, O., Lenz, R. & Sharma, M. (1999). Emission of
reactive terpene compounds from orange orchards and their removal by within-
canopy processes, J. Geophys. Res., Vol. 104, pp. 8077–8094, 01480227
160 Metabolomics

Claus, S.P., Tsang, T.M., Wang, Y., Cloarec, O., Skordi, E., Martin, F.P., Rezzi, S., Ross, A.,
Kochhar, S., Holmes, E. & Nicholson, J.K. (2008). Systemic multicompartmental
effects of the gut microbiome on mouce metabolic phenotypes. Molecular Systems
Biology, Vol. 4, No. 219, pp. 1-14, 17444292
Clausing, P. & Gottschalk, M. (1989). Effects of drinking water acidification, restriction of
water supply and individual cageing on parameters of toxicological studies in rats.
Z. Versuchstierkd., Vol. 32, No 3, pp. 129–134, 00443697
Czerniak, R. (2001). Gender-based differences in pharmacokinetics in laboratory animal
models. Int. J. Toxicol., Vol. 20, No. 3, pp. 161–163, 10915818
Davison, J.M. & Hytten, F.E. (1975). The effect of pregnancy on the renal handling of
glucose. Br. J. Obstet. Gynaecol., Vol. 82, No 5, pp. 374–381, 03065456
Dudareva, N., Murfitt, L.M., Mann, C.J., Gorenstein, N., Kolosova, N., Kish, C.M., Bonham,
C. & Wood, K. (2000). Developmental regulation of methyl benzoate biosynthesis
and emission in snapdragon flowers. Plant Cell, Vol. 12, pp. 949-961, 10404651
Duhl, T.R., Helmig, D. & Guenther, A. (2008). Sesquiterpene emissions from vegetation: a
review. Biogeosciences, Vol. 5, pp. 761–777, 17264170
Duncan, S.H., Belenguer, A., Holtrop, G., Johnstone, A.M., Flint, H.J. & Lobley, G.E. (2007).
Reduced Dietary Intake of Carbohydrates by Obese Subjects Results in Decreased
Concentrations of Butyrate and Butyrate-Producing Bacteria in Feces. Appl. Environ.
Microbiol.,Vol. 73, No. 4, pp. 1073-1078, 00992240
Dunne, C. (2001). Adaptation of bacteria to the intestinal niche: probiotics and gut disorder.
Inflamm. Bowel. Dis., Vol. 7, pp. 136–145, 10780998
Felgines, C., Talavéra, S., Texier, O., Gil-Izquierdo, A., Lamaison, J.-L. & Rémésy, C. (2005).
Blackberry anthocyanins are mainly recovered from urine as methylated and
glucuronidated conjugates in humans. J. Agric. Food Chem., Vol. 53, pp. 7721–7727,
00218561
Freese, R., Alfthan, G., Jauhiainen, M., Basu, S., Erlund, I., Salminen, I., Aro, A. & Mutanen,
M. (2002). High intakes of vegetables, berries, and apples combined with a high
intake of linoleic or oleic acid only slightly affect markers of lipid peroxidation and
lipoprotein metabolism in healthy subjects. Am. J. Clin. Nutr., Vol. 76, pp. 950-960,
00029165
Gaillard, Y., Vayssette, F., Balland, A., Pépin, G. (1999). Gas chromatographic-tandem mass
spectrometric determination of anabolic steroids and their esters in hair:
application in doping control and meat quality control. J. Chromatogr. B, Vol. 735,
pp. 189-205, 15700232
Gattuso, G., Barreca, D., Gargiulli, C., Leuzzi, U. & Caristi, C. (2007). Flavonoid Composition
of Citrus Juices. Molecules, Vol. 12, pp. 1641-1673, 14203049
Gavaghan, C.L., Holmes, E., Lenz, E., Wilson, I.D. & Nicholson, J.K. (2000). An NMR-based
metabonomic approach to investigate the biochemical consequences of genetic
strain differences: application to the C57BL10J and Alpk:ApfCD mouse. FEBS Lett.,
Vol. 484, No. 3, pp. 169–174, 00145793
Gouinguené, S. P. & Turlings, T. C. J. (2002). The effects of abiotic factors on induced volatile
emissions in corn plants. Plant Phys., Vol. 129, pp. 1296–1307, 00320889
Groopman, J.D., Wogan, G.N., Roebuck, B.D. & Kensler, T.W. (1994). Molecular biomarkers
for aflatoxins and theirapplication to human cancer prevention. Cancer Res. Vol. 54,
pp. 1907-1911, 00085472
Metabotype Concept: Flexibility, Usefulness and Meaning in Different Biological Populations 161

Hakola, H., Laurila, T., Lindfors, V., Hellén, H., Gaman, A., & Rinne, J. (2001). Variation of
the VOC emission rates of birch species during the growing season. Boreal Environ.
Res., Vol. 6, pp. 237–249, 12396095
Hamilton, H.E., Wallace, J.E., Shimek E.L. Jr, Land, P., Harris, S.C. & Christenson J.G. (1977).
Cocaine and benzoylecgonine excretion in humans. J. Forensic Sci., Vol. 22, No. 4,
pp. 697-707, 00221198
Hansen, U. & Seufert, G. (1999). Terpenoid emission from Citrus sinensis (L.) OSBECK under
drought stress. Phys. Chem. Earth (B), Vol. 24, pp. 681–687, 14747065
Harborne, J.B. (1966b). Caffeic acid Ester distribution in higher plants. Z. Naturforsch. 21b,
604-605.
Hemminki, K. & Vodicka, P. (1995). Styrene: from characterization of DNA adducts to
application in styrene-exposed lamination workers. Toxicol. Lett., Vol. 77, pp. 153-
161, 03784274
Hodgkinson, A. (1962). Citric acid excretion in normal adults and in patients with renal
calculus. Clin. Sci., Vol. 23, pp. 203–205, 01435221
Holden, J.M., Bhagwat, S.A., Haytowitz, D.B., Gebhardt, S.E., Dwyer, J.T., Peterson, J.,
Beecher, G.R., Eldridge, A.L. & Balentine, D. (2005). Development of a database of
critically evaluated flavonoids data: application of USDA’s data quality evaluation
system. J. Food Comp. Anal., Vol. 18, pp. 829–844, 08891575
Holmes, E., Bonner, F.W. & Nicholson, J.K. (1995). Comparative studies on the
nephrotoxicity of 2-bromoethanamine hydrobromide in the Fischer 344 rat and the
multimammate desert mouse (Mastomys natalensis). Arch. Toxicol., Vol. 70, No 2, pp.
89–95, 03405761
Holmes, E., Bonner, F.W. & Nicholson, J.K. (1996). Comparative biochemical effects of low
doses of mercury II chloride in the F344 rat and the Multimammate mouse
(Mastomys natalensis). Comp. Biochem. Physiol., Vol. 114C, No. 1, pp. 7–15, 15320456
Holmes, E., Bonner, F.W. & Nicholson, J.K. (1997). 1H NMR spectroscopic and
histopathological studies on propyleneimine-induced renal papillary necrosis in
the rat and the multimammate desert mouse (Mastomys natalensis). Comp. Biochem.
Physiol. C. Pharmac. Toxicol. Endocrinol., Vol. 116, No. 2, pp. 125–134, 07428413
Holmes, E., Nicholls, A.W., Lindon, J.C., Ramos, S., Spraul, M., Neidig, P., Connor, S.C.,
Connelly, J., Damnent, S.J., Haselden, J. & Nicholson, J.K. (1998). Development of a
model for classification of toxin-induced lesions using 1H NMR spectroscopy of
urine combined with pattern recognition. NMR Biomed., Vol. 11, No. 4–5, pp. 235–
244, 09523480
Holmes, E., Nicholls, A.W., Lindon, J.C., Connor, S.C., Connelly, J.C., Haselden, J.N.,
Damment, S.J., Spraul, M., Neidig, P. & Nicholson, J.K. (2000). Chemometric
models for toxicity classification based on NMR spectra of biofluids. Chem. Res.
Toxicol., Vol. 13, No. 6, pp. 471–478, 0893228X
Holmes, E., Wilson, I.D. & Nicholson, J.K. (2008). Metabolic Phenotyping in Health and
Disease. Cell, Vol. 134, pp. 714-717, 00928674
Hugget, R.J., Kimerle, R.A., Mehrle, P.M. & Bergman, H.L. (1992). Biomarkers. Biochemical,
Physiological and Histological Markers of Anthropogenic Stress. Lewis, Chelsea,
MI. ISBN: 978-0873715058.
Iwashina, T. (2000). The structure and distribution of the flavonoids in plants. J. Plant Res.,
Vol. 113, pp. 287-299, 09189440
162 Metabolomics

Janicsák, G., Máthé, I., Miklóssy-Vári, V. & Blunden, G. (1999). Comparative studies of the
rosmarinic and caffeic acid contents of Lamiaceae species. Biochem. Syst. Ecol., Vol.
27, pp. 733-738, 03051978
Jones, A.R. (1982). Some observations on the urinary excretion of glycine conjugates by
laboratory animals. Xenobiotica, Vol. 12, pp. 387–395, 00498254
Joossen, J.V. (1988). Mechanisms of hypercholesterolemia and atherosclerosis. Acta Cardiol.
Suppl. Vol. 29, pp. 63–83
Kaufman, P.B., Duke, J.A., Brielmann, H., Boik, J. & Hoyt, J.E. (1997). A Comparative Survey
of Leguminous Plants as Sources of the Isoflavones, Genistein and Daidzein:
Implications for Human Nutrition and Health. The Journal of Alternative and
Complementary Medicine, Vol. 3, No. 1, pp. 7-12, 10755535
Kawaii, S., Tomono, Y., Katase, E., Ogawa, K. & Yano, M. (1999). Quantitation of flavonoid
constituents in citrus fruits. J. Agric. Food Chem., Vol. 47, No. 9, pp. 3565-3571,
00218561
Kidd, P.S., Llugany, M., Poschenrieder, C., Gunsé, B & Barceló, J. (2001). The role of root
exudates in aluminium resistance and silicon-induced amelioration of aluminium
toxicity in three variety of maize (Zea mays L.). J Exp Bot, Vol. 52, pp. 1959-1967,
00220957
Kintz, P., Cirimele, V., Jeanneau, T. & Ludes, B. (1999). Identification of testosterone and
testosterone esters in human hair. J. Anal. Toxicol., Vol. 23, pp. 352-356, 01464760
Konishi, H., Tanaka, K., Minouchi, T. & Yamaji, A. (2004). Urinary 6β-hydroxycortisol/17-
hydroxycorticosteroids ratio as a measure of hepatic CYP3A4 capacity after
enzyme induction. Ann Clin Biochem, Vol. 41, pp. 335-337, 00045632
Krol, M., Gray, G.R., Hurry, V.M., Oquist, G., Malek, L. & Huner N.P.A. (1995). Low-
temperature stress and photoperiod affect an increased tolerance to photoinhibition
in Pinus banksiana seedlings. Can. J. Bot., Vol. 73, pp. 1119-1127, 00084826
Kühnau, J. (1976). The flavonoids. A class of semi-essential food components: their role in
human nutrition. World Rev. Nutr. Diet., Vol. 24, pp. 117-191, 00842230
Leyva A., Jarillo J. A., Salinas J. & Martinez-Zapater J.M. (1995). Low temperature induces
the accumulation of phenylalanine ammonia-lyase and chalcone synthase mRNAs
of Arabidopsis thaliana in a light-dependent manner. Plant Physiol., Vol. 108, pp. 39-
46, 00320889
Liggins, J., Bluck, L.J., Runswick, S., Atkinson, C., Coward, W.A. & Bingham, S.A. (2000).
Daidzein and genistein contents of vegetables. Br. J. Nutr., Vol. 84, pp. 717- 25,
00071145
Logue, B.A., Kirschten, N.P., Petrikovics I., Moser, M.A., Rockwood, G.A., Baskin, S.I. (2005).
Determination of the cyanide metabolite 2-aminothiazoline-4-carboxylic acid in
urine and plasma by gas chromatography-mass spectrometry. J Chromatogr B, Vol.
819, No. 2, pp. 237-244, 15700232
Lutz, U., Bittner, N., Ufer, M. & Lutz, W. K. (2010). Quantification of cortisol and 6 beta-
hydroxycortisol in human urine by LC MS/MS, and gender-specific evaluation of
the metabolic ratio as biomarker of CYP3A activity. J. Chromatogr. B, Vol. 878, No. 1,
pp. 97–101, 15700232
Majewska, M. Skrzycki, M., Podsiad, M. & Czeczot, H. (2011). Evaluation of antioxidant
potential of flavonoids: an in vitro study. Acta Poloniae Pharmaceutica. Drug Research,
Vol. 68, No 4, pp. 611-615, 00016837
Metabotype Concept: Flexibility, Usefulness and Meaning in Different Biological Populations 163

Maslowski, K.M., Vieira, A.T., Ng, A., Kranich, J., Sierro, F., Yu, D., Schilter, H.C., Rolph,
M.S., Mackay, F., Artis, D., Xavier, R.J., Teixeira, M.M. & Mackay, C.R. (2009).
Regulation of inflammatory responses by gut microbiota and chemoattractant
receptor GPR43. Nature, Vol. 461, pp. 1282-1286, 00280836
Mayr, M., Yusuf, S., Weir, G., Chung, Y.L., Mayr, U., Yin, X., Ladroue, C., Madhu, B.,
Roberts, N., De Souza, A., Fredericks, S., Stubbs, M., Griffiths, J.R., Jahangiri, M.,
Xu, Q. & Camm, A.J. (2008). Combined metabolomic and proteomic analysis of
human atrial fibrillation. Journal of the American College of Cardiology, Vol. 51, No. 5,
pp. 585–594, 07351097
Kim, Y.S., & Milner, J.A. (2011). Bioactive Food Components and Cancer-Specific
Metabonomic Profiles. Journal of Biomedicine and Biotechnology ID 721213, pp. 1-9,
11107243
Mazurek, S. (2011). Pyruvate kinase type M2: a key regulator of the metabolic budget system
in tumor cells. Int J Biochem Cell Biol., Vol. 43, No. 7, pp. 969-980, 0020711X
Moreno, A. & Arus, C. (1996). Quantitative and qualitative characterization of 1H NMR
spectra of colon tumors, normal mucosa and their perchloric acid extracts:
decreased levels of myo-inositol in tumours can be detected in intact biopsies. NMR
Biomed., Vol. 9, pp. 33–45, 09523480
Mouly, P., Gaydou, E. M. & Auffray, A. (1998). Simultaneous separation of flavanone
glycosides and polymethoxylated flavones in citrus juices using liquid
chromatography. J. Chromatogr., Vol. 800, No. 2, pp. 171–179, 0021-9673.
Mulder, T.P., Rietveld, A.G. & van Amelsvoort, J.M. (2005). Consumption of both black tea
and green tea results in an increase in the excretion of hippuric acid into urine. Am.
J. Clin. Nutr., Vol. 81, pp. 256S–260S, 00029165
Mullen, W., Edwards, C.A. & Crozier, A. (2006). Absorption, excretion and metabolic
profiling of methyl-, glucuronyl-, glucosyl and sulpho-conjugates of quercetin in
human plasma and urine after ingestion of onions. Br. J. Nutr., Vol. 96, pp. 107–116,
00071145
Mullen, W., Edwards, C.A., Serafini, M., Crozier, A. (2008). Bioavailability of pelargonidin-3-
O-glucoside and its metabolites in humans following the ingestion of strawberries
with and without cream. J. Agric. Food Chem., Vol. 56, pp. 713–719, 00218561
Nardini, M.E., Cirillo, F., Natella, C. & Scaccini, C. (2002). Absorption of phenolic acids in
humans after coffee consumption. J. Agric. Food Chem., Vol. 50, pp. 5735–5741,
00218561
Oren-Shamir, M. & Levi-Nissim, A. (1997). Temperature effect on the leaf pigmentation of
Cotinus coggygria 'Royal Purple'. J. Hor. Sci. Biotech., Vol. 72, pp. 425-432. 14620316
Ormeño, E., Mévy, J.P., Vila, B., Bousquet-Mélou, A., Greff, S., Bonin, G. & Fernandez, C.
(2007a). Water deficit stress induces different monoterpene and sesquiterpene
emission changes in Mediterranean species. Relationship between terpene
emissions and plant water potential. Chemosphere, Vol. 67, pp. 276-284, 00456535
Ormeño, E., Fernandez, C., Bousquet-Mélou, A., Greff, S., Morin, E., Robles, C., Vila, B. &
Bonin, G. (2007b). Monoterpene and sesquiterpene emissions of three
Mediterranean species through calcareous and siliceous soils in natural conditions,
Atmos. Environ., Vol. 41, pp. 629-639, 13522310
164 Metabolomics

Phipps, A.N., Wright, B., Stewart, J. & Wilson, I.D. (1997). Use of proton NMR for
determining changes in metabolite excretion profiles induced by dietary changes in
the rat. Pharmacy and Pharmacology Communications, Vol. 3, pp. 143–146, 20427158
Platz, E.A., Till C., Goodman, P.J., Parnes, H.L., Figg, W.D., Albanes,D., Neuhouser, M.L.,
Klein, E.A., Thompson, I.M., Jr & Kristal, A.R. (2009). Men with low serum
cholesterol have a lower risk of high-grade prostate cancer in the placebo arm of
the prostate cancer prevention trial. Cancer Epidemiol Biomarkers Prev, Vol. 18, pp.
2807–2813, 10559965
Prevost, V., Likhachev, A.J., Loktionova, N.A., Bartsch, H., Wild, C.P., Kazanova, O.I.,
Arkhipov, A.I., Gershanovich, M.L. & Shuker, D.E.G. (1996). DNA base adducts in
urine and white blood cells of cancer patients receiving combination
chemotherapies which include N-methyl-N-nitrosourea. Biomarkers, Vol. 1, pp. 244-
251, 1354750X
Radtke, J., Linseisen, J. & Wolfram, G. (2002). Fasting plasma concentrations of selected
flavonoids as markers of their ordinary dietary intake. Eur. J. Nutr., Vol. 41, pp.203-
209, 14366207
Raina, K., Serkova, N. J. & Agarwal, R. (2009). Silibinin feeding alters the metabolic profile
in TRAMP prostatic tumors:1hnmrs-based metabolomics study. Cancer Research,
Vol. 69, No. 9, pp. 3731–3735, 00085472
Rechner, A.R., Spencer, J.P.E., Kuhnle, G., Hahn, U. & Rice-Evans, C.A. (2001). Novel
biomarkers of the metabolism of caffeic acid derivatives in vivo. Free Rad. Biol.
Med., Vol. 30, pp. 1213–1222, 08915849
Rikimaru, T., Oozeki, T., Ichikawa, M., Ebisawa, H. & Fujita, Y. ( 1989). Comparisons of
urinary creatinine, skeletal muscle mass and indices of muscle protein catabolism
in rats fed ad libitum, with restricted food intake and deprived of food. J. Nutr. Sci.
Vitaminol., Vol. 35, No. 3, pp. 190–209, 03014800
Robertson, D.G., Reily, M.D., Lindon, J.C., Holmes, E. & Nicholson, J.K. (2002).
Metabonomic technology as a tool for rapid throughput in vivo toxicity screening.
In Comprehensive Toxicology Vol. XIV: Molecular and Cellular Toxicology, Heuvel
JPV, Perdew GH, Mattes WB & Greenlee WF (Eds). Elsevier (2002). pp. 583-610,
9780080468846, Amsterdam
Roitto, M., Rautio, P., Julkunen-Tiitto, R., Kukkola, E. & Huttunen, S. (2005). Changes in the
concentrations of phenolics and photosynthates in Scots pine (Pinus sylvestris L.)
seedlings exposed to nickel and copper. Environmental Pollution, Vol. 137, pp. 603-
609, 02697491
Schwiertz, A., Taras, D., Schäfer, K., Beijer, S., Bos, N.A., Donus, C. & Hardt, P.D. (2010).
Microbiota and SCFA in lean and overweight healthy subjects. Obesity, Vol. 18, pp.
190-195, 19307381
Semmar, N., Bruguerolle, B., Boullu-Ciocca, S. & Simon, N (2005a). Cluster Analysis: An
Alternative Method for Covariate Selection in population Pharmacokinetic
Modeling. Journal of Pharmacokinetics and Pharmacodynamics, Vol. 32, No. 3-4, pp.
333-358, 1567567X
Semmar, N., Jay, M., Farman, M. and Chemli, R. (2005b). Chemotaxonomic analysis of
Astragalus caprinus (Fabaceae) based on the flavonic patterns. Biochem. Syst. Ecol.,
Vol. 33, pp. 187-200, 03051978
Metabotype Concept: Flexibility, Usefulness and Meaning in Different Biological Populations 165

Semmar, N., Jay M. & Nouira S. (2007). A new approach to graphical and numerical analysis
of links between plant chemotaxonomy and secondary metabolism from HPLC
data smoothed by a simplex mixture design. Chemoecology, Vol. 17, No. 3, pp. 139-
156, 09377409
Semmar, N., Nouira, S. & Farman, M. (2008). Variability and Ecological Significance of
Secondary Metabolites in Terrestrial Plant World. In: Handbook of Nature
Conservation, Aronoff, J.B., pp. 1-89. Nova Science Publishers, 9781606929933, NY
Semmar, N. (2010). Chemotaxonomical Analysis of Herbaceous Plants Based on Phenolic and
Terpenic patterns: flexible tools to survey biodiversity in grassland. Nova Science
Publishers, 9781616687892, NY.
Shibata, M. & Ishikura, N. (1960). Paper chromatographic survey of anthocyanin in tulip-
flowers. I. Jap. J. Bot., Vol. 17, pp. 230-238
Stanley, E.G. (2002). 1H NMR spectroscopic and chemometric studies on endogenous
physiological variation in rats. Ph.D. thesis, university of London, pp. 43–67
Tarvainen, V., Hakola, H., Hellén, H., Bäck, J., Hari, P. & Kulmala, M. (2005): Temperature
and light dependence of the VOC emissions of Scots Pine, Atmos. Chem. Phys., Vol.
5, pp. 989–998, 16807367
Thieme, D., Grosse, J., Sachs, H. & Mueller, R.K. (2000). Analytical strategy for detecting
doping agents in hair. Forensic Sci. Int., Vol. 107, pp. 335-345, 03790738
Thysell, E., Surowiec, I., Hörnberg, E., Crnalic, S., Widmark, A., Johansson, A.I., Stattin, P.,
Bergh, A., Moritz, T., Antti., H. & Wikström, P. (2010). Metabolomic
Characterization of Human Prostate Cancer Bone Metastases Reveals Increased
Levels of Cholesterol. PLoS One, Vol. 5, No. 12, e14175, 19326203
Timbrell, J.A., Draper, R.P. & Waterfield, C.J. (1994). Biomarkers in toxicology: new uses for
some old molecules? Toxicol. Ecotoxicol. News, Vol. 1, pp. 4-14, 13504592
Timbrell, J.A. (1998). Biomarkers in toxicology. Toxicology, Vol. 129, pp. 1–12, 0300483X
Tomás-Barberán, F.A.T. & Wollenweber, E. (1990). Flavonoid aglycones from the leaf
surfaces of some Labiatae species. Pl. Syst Evol., vol. 173, pp. 109-118, 03782697
Torskangerpoll, K., Nørbæk, R., Nodland, E., Øvstedal, D.O. & Andersen, Ø.M. (2005).
Anthocyanin content of Tulipa species and cultivars and its impact on tepal
colours. Biochem. Syst. Ecol., Vol. 33, pp. 499-510, 03051978
Turer, A. T., Stevens, R. D., Bain, J. R., Muehlbauer, M.J., van der Westhuizen, J., Mathew,
J.P., Schwinn, D.A., Glower, D.D., Newgard, C.B. & Podgoreanu M.V. (2009).
Metabolomic profiling reveals distinct patterns of myocardial substrate use in
humans with coronary artery disease or left ventricular dysfunction during surgical
ischemia/reperfusion. Circulation, Vol. 119, No. 13, pp. 1736–1746, 00097322
Turlings, T. C. J., Loughrin, J. H., McCall, P. J., R¨ose, U. S. R., Lewis, W. J., & Tumlinson, J.
H. (1995). How caterpillar-damaged plants protect themselves by attracting
parasitic wasps, Proc. Natl. Acad. Sci., Vol. 92, pp. 4169–4174, 00278424
Vallejo, M., Garcia, A., Tunon, J. , García-Martínez, D., Angulo, S., Martin-Ventura, J.L.,
Blanco-Colio, L.M., Almeida, P., Egido, J. & Barbas, C. (2009). Plasma fingerprinting
with GC-MS in acute coronary syndrome. Analytical and Bioanalytical Chemistry,
Vol. 394, No. 6, pp. 1517–1524, 16182642
van Eijk, J.P., Nieuwhof, M., van Keulen, H.A. & Keijzer, P. (1987). Flower colour analyses in
tulip (Tulipa L.). The occurrence of carotenoids and flavonoids in tulip tepals.
Euphytica, Vol. 36, pp. 855-862, 00142336
166 Metabolomics

Van Welie, R.T.H., van Dijck, R.G.J.M., Vermeulen, N.P.E., van Sittert, N.J., 1992.
Mercapturic acids, protein adducts and DNA adducts as biomarkers of
electrophilic chemicals. Crit. Rev. Toxicol., Vol. 22, No. 5-6, pp. 271-306, 10408444
Williams, C.A. (1975). Biosystematics of the Monocotyledoneae – Flavonoid Patterns in
Leaves of the Liliaceae. Biochem. Syst. Ecol., Vol. 3, pp. 229-244, 03051978
Wilson, I.D. (2009). Drugs, bugs, and personalized medicine: Pharmacometabonomics enters
the ring. PNAS, Vol. 106, No. 34, pp. 14187-14188, 00278424
Xu, X., Harris, K.S., Huei-Ju, W., Murphy, P.A. & Hendrich, S. (1995). Bioavailability of
soybean isoflavones depends upon gut microflora in women. The Journal of
Nutrition, Vol. 125, No. 9, pp.2307-2315, 00223166
Yanez, J.A., Remsberg, C.M., Miranda, N.D., Vega-Villa, K.R., Andrews, P.K. & Davies,
N.M. (2008). Pharmacokinetics of Selected Chiral Flavonoids: Hesperetin,
Naringenin and Eriodictyol in Rats and their Content in Fruit Juices. Biopharm.
Drug Dispos., Vol. 29, pp. 63–82, 01422782
Yang, C., Richardson A.D., Smith, J.W. & Osterman A. (2007). Comparative metabolomics of
breast cancer. Pacific Symposium on Biocomputing 12, 181-192, 17935091
Zhang, A.Q., Mitchell, S.C. & Smith, R.L. (1996). Exacerbation of symptoms of fish-odour
syndrome during menstruation. Lancet, Vol. 348, No. 9043, pp. 1740–1741, 01406736
Zheng, W. & Wang, S.Y. (2003). Oxygen radical absorbing capacity of phenolics in
blueberries, cranberries, chokeberries, and lingonberries. J. Agric. Food Chem., Vol.
51, pp. 502-509, 00218561
Zoetendal, E.G., Rajilic-Stojanovic, M. & de Vos, W.M. (2008). High throughput diversity
and functionality analysis of the gastrointestinal tract microbiota. Gut., Vol. 57, pp.
1605-1615, 00175749
7

Software Techniques for Enabling


High-Throughput Analysis of
Metabolomic Datasets
Corey D. DeHaven, Anne M. Evans, Hongping Dai and Kay A. Lawton
Metabolon, Inc.
United States of America

1. Introduction
In recent years, the study of metabolomics and the use of metabolomics data to answer a
variety of biological questions have been greatly increasing (Fan, Lane et al. 2004; Griffin
2006; Khoo and Al-Rubeai 2007; Lindon, Holmes et al. 2007; Lawton, Berger et al. 2008).
While various techniques are available for analyzing this type of data (Bryan, Brennan et al.
2008; Scalbert, Brennan et al. 2009; Thielen, Heinen et al. 2009; Xia, Psychogios et al. 2009),
the fundamental goal of the analysis is the same – to quickly and accurately identify
detected molecules so that biological mechanisms and modes of action can be understood.
Metabolomics analysis was long thought of as, and in many aspects still is, an
instrumentation problem; the better and more accurate the instrumentation (LC/MS,
GC/MS, NMR, CE, etc.) the better the resulting data which, in turn, facilitates data
interpretation and, ultimately, the understanding of the biological relevance of the results.
While the quality of instrumentation does play a very important role, the rate-limiting step
is often the processing of the data. Thus, software and computational tools play an
important and direct role in the ability to process, analyze, and interpret metabolomics data.
This situation is much like the early days of automated DNA sequencing where it was the
evolution of the software components from highly manual to fully automated processes that
brought about significant advances and a new era in the technology (Hood, Hunkapiller et
al. 1987; Hunkapiller, Kaiser et al. 1991; Fields 1996). Currently, software tools exist for the
automated initial processing of metabolomic data, especially chromatographic separation
coupled to mass spectrometry data (Wilson, Nicholson et al. 2005; Nordstrom, O'Maille et al.
2006; Want, Nordstrom et al. 2007; Patterson, Li et al. 2008). Samples can be processed
automatically; peak detection, integration and alignment, and various quality control (QC)
steps on the data itself can be performed with little to no user interaction. However, the
problem is that the generation of data, together with peak detection and integration, is the
relatively simple part; without a properly engineered system for managing this part of the
process the vast number of data files generated can quickly become overwhelming.
Two major processes in metabolomic data processing are the verification of the accuracy of
the peak integration and the verification of the accuracy of the automated identification of
the metabolites that those peaks represent. These two processes, while vitally important to
168 Metabolomics

the accuracy of the results, are very time consuming and are the most significant bottlenecks
in processing metabolomic data. In fact, the peak integration verification step is often
omitted due to the extremely large number of peaks whose integration must be verified.

2. Background
At the outset, running a metabolomics study is actually simple and straightforward.
Samples are prepared for running on a signal detection platform, signal data is collected on
samples from the instrumentation, the signals are translated into peaks, the peaks are
compared to reference libraries for the identification of metabolites and those identified
metabolites are then statistically analyzed with whatever metadata may exist for the
samples. Alternatively, the entirety of the detected peaks resulting from the instrument
signal data are statistically analyzed without metabolite identification prior to the statistical
analysis.
Once statistical analysis is completed and the significant signals have been stratified and
metabolites identified, biochemical pathway analysis is performed to gain insight into the
original biological questions the study asked. Too often, when the metabolomic experiments
do not provide meaningful biological results, the realization may come that there’s so much
variability in the data, it can’t be used to address the original objectives of the study. Despite
the methods and software provided by the various instrument vendors, it turns out that
running a global, non-targeted analysis of small molecules in a complex mixture that
generates high-quality data and provides answers to biological questions is challenging.
Doing so in a high-throughput environment is significantly more challenging.
However, a high-throughput metabolomics platform that produces reliable, precise,
reproducible, and interpretable data is possible. It simply requires the right process coupled
with the right software tools. As with any high throughput process it is important to have a
logical, consistent workflow that is simple, reproducible, and expandable without
negatively impacting the efficiency of the process. It is important to know when human
interaction is required and when it is not. Well designed and integrated software can
efficiently handle the majority of the mundane workload, allowing human interaction to be
focused only where required.

3. Approach
Metabolite identification is essential for chemical and biological interpretation of
metabolomics experiments. Two approaches to metabolomic data analysis have been used
and will be described in detail below. The main difference between the two approaches is
when the metabolite is identified, either before or after statistical analysis of the data.
To date, the most commonly used method of processing metabolomic data has been to
statistically analyze all of the detected ion-features (‘ion-centric’). Ion features, defined here
as a chromatographic peak with a given retention time and m/z value, are analyzed using a
statistical package such as SAS or S-plus to determine which features vary statistically
significantly and are related to a test hypothesis (Tolstikov and Fiehn 2002; Katajamaa and
Oresic 2007; Werner, Heilier et al. 2008) . The significant ion feature changes are then used to
prioritize metabolite identification. One issue with this type of approach is the convoluted
Software Techniques for Enabling High-Throughput Analysis of Metabolomic Datasets 169

nature of the data being analyzed. In many cases the “statistically significant ion-features”
are various forms of the same chemical and are therefore redundant information. Most
biochemicals detected in a traditional LC- or GC-MS analysis produce several different ions,
which contributes to the massive size and complexity of metabolomics data. In addition,
there are an even larger number of measurements for each experimental sample which
impacts the false discovery rate (Benjamini and Hochberg 1995; Storey and Tibshirani 2003).
In the ‘chemo-centric’ approach to metabolomics data analysis discussed here, metabolites
are identified on the front-end through the use of a reference library comprised of spectra of
authentic chemical standards(Lawton, Berger et al. 2008; Evans, Dehaven et al. 2009). Then,
instead of treating all detected peaks independently, as is done in the ion-centric approach,
the chemo-centric method selects a single ion (‘quant-ion’) to represent that metabolite in all
subsequent analyses. The other ions associated with the metabolite are essentially
redundant information that only add to data complexity. Furthermore, the statistical
analysis may be skewed since a single metabolite may be represented by multiple ion peaks,
and the false discovery rate increased due to the large number of measurements relative to
the number of samples in the experiment. Accordingly, by taking a chemo-centric approach
any extraneous peaks can be identified and removed from the analysis based on the
authentic standard library/database. Since the number of features analyzed statistically
contributes to the probability of obtaining false positives, analyzing one representative ion
for each metabolite reduces the number of false positives. Further, the chemo-centric data
analysis method is powerful because a significant amount of computational processing time
and power can be saved simply due to data reduction.
The majority of work and complexity with the chemo-centric approach are: first, the
generation of the reference library of spectra from authentic chemical standards; second, the
actual identification of the detected metabolites using the reference library; and third, the
ability for quality control (QC) of the automated metabolite identification, peak detection
and integration. Notably, the QC of the automated processes is often overlooked. However,
the QC step is critical to ensure that false identifications and poor or inconsistent peak
integrations do not make their way into the statistical analysis of the experimental results.
The generation of a reference library entry made up of the spectral signature and
chromatographic elution time of an authentic chemical standard is relatively
straightforward, as is the generation of spectral-matching algorithms that use the reference
library to identify the experimentally detected metabolites. In contrast, performing the QC
step on the automated processes, including peak detection, integration and metabolite
identification, is time and human resource intensive.
Not to be overlooked, an issue with using a reference library comprised of authentic
standards is dealing with metabolites in the samples that are not contained within the
reference library. The power of the technology would be significantly reduced if it was
limited to identifying only compounds contained in the reference library. Through
intelligent software algorithms, it is possible to analyze data of similar characteristics across
multiple samples in a study to find those metabolites that are unknown by virtue of not
matching a reference standard in the library, and, in the process, group all the ion-features
related to that unknown together by examining ion correlations across the sample set
(Dehaven, Evans et al. 2010). One such method capitalizes on the natural biological
variability inherent in the experimental samples, using this variation the metabolites and
170 Metabolomics

their respective ion-features can reveal themselves and be entered into the chemical
reference library as a novel chemical entity (Dehaven, Evans et al. 2010). The unknown
chemical can then be tracked in future metabolomics studies, and, if important, can be
identified using standard analytical chemistry techniques.
Without going into detail, it is important to note that the sample preparation process is
critical. High quality samples that have been properly and consistently prepared for analysis
on sensitive scientific instrumentation are of extreme importance. Ensuring this high quality
starts with the collection and preparation of the samples. No software system is going to be
able to produce high-quality data unless ample effort is focused on consistently following
standardized protocols for preparing high quality samples for analysis.
The following discussion, examples and workflow solutions make use of GCMS or LCMS
(or both) platforms for metabolomic analysis of samples, although the concepts in general
could apply to a variety of data collection techniques. Software tools are also presented to
demonstrate the application of the concepts that are discussed but the tools themselves will
not be discussed in great detail. It is also important to note that achieving the greatest
operation efficiency of the process relies on treating all of the experimental samples in a
study as a set and not as individual files. By using tools to analyze and perform quality
control on the samples as a single group or set it becomes much easier to spot patterns that
can be useful to determine what is going on in the overall process.

4. Processing data files, peak detection, alignment, and metabolite


identification
4.1 File processing can become a major hurdle
There is no shortage of software available on the market to read spectral data and detect the
start and the stop of peaks, and the baseline, and then calculate the area inside of those
peaks. Each instrument vendor provides some flavor of detection and analysis software
with their instrument and several open-source and commercial efforts to read spectral data
and produce integrated peak data regardless of vendor format are available (Tolstikov and
Fiehn 2002; Katajamaa and Oresic 2005; Katajamaa and Oresic 2007). In almost all cases,
these packages do a complete job of finding and integrating peaks and do so in a reasonable
amount of time. Thus, the peak detection and integration process is not the rate-limiting step
when it comes to data quality and automated processing.
As it turns out, the file processing problem is primarily a file management problem that is
the result of two issues – human and machine. The first problem stems from human
interaction, in that a human being can introduce more error and inconsistency than is
acceptable. Optimally, a human should play no role in the naming or processing of
instruments data files. Naming of instrument data files should take place within the system
used to track sample information, a LIMS for instance. The LIMS or other sample tracking
system should generate a sample list and run order for the samples to be run on the
instrument using a consistent naming convention that can be easily associated with the
sample in question. The second problem stems from both machine and human. The software
performing the peak detection and integration must have the capability of automatically
processing a data file when presented with it, then archiving the file when completed. And,
Software Techniques for Enabling High-Throughput Analysis of Metabolomic Datasets 171

in high-throughput mode, it is best not to have humans manage data files, either in storage
locations, or, as noted above, in naming. For consistency, it is imperative that the machines
control this step; running one experiment on one machine may be manageable manually but
running experiments in tandem or on more than one instrument can easily result in
misnaming, file version problems, location mishaps, etc. if file management is not
automated.

4.2 Manual integration of peak data is inadequate for high-throughput processing


Processing metabolomics data in a high-throughput setting requires automated processing
of data files. While an SDK (software development kit) is provided by many instrument
vendors, and there are commercial and open source packages for creating this functionality
available (Smith, Want et al. 2006), not all vendor software permits this functionality. One of
the main reasons automated peak integration works well is because it allows data to be
rapidly uploaded and processed. Manual integration, while perhaps more accurate,
dramatically slows the peak analysis process. Further QC and refinement of the automated
peak integration can be performed more optimally later in the process, where, in practice,
the bar for peak detection can be slightly lower. The reasons that the bar for peak detection
can be reduced will be discussed below.

4.3 Alignment based on peak similarity inadequate, retention index should be used
Many of the software packages provide capabilities to align the chromatograms to
account for time drift in an instrument. In many instances internal standards and/or
endogenous metabolites are used across the analyzed samples to align chromatography
based on their retention times, such that there is confidence that the same peak at the
same mass is consistent among the data files. This approach should be avoided because
while it works fine for peak analysis and chromatographic alignment on a single, small
study it will only be applicable within that one study where retention times are quite
consistent. This type of alignment approach makes it much harder to do a comparison to a
reference standard library where a retention profile is used as matching criteria. The
better choice is to opt for retention index (RI) calculation, which can correctly align
chromatograms even over long periods of time where conditions can be vastly different
dependent on the condition in these systems. Using a retention index method, each RT
marker is given a fixed RI value (Evans, Dehaven et al. 2009). The retention times for the
retention markers can be set in the integrator method and the time at which those internal
standards elute are used to calculate an adjustment RI ladder. All other detected peaks
can then use their actual retention time and adjustment index to calculate a retention
index. In this way, all detected peaks are aligned based on their elution relative to their
flanking RT markers. An RI removes any systematic changes in retention time by
assuming that the compound will always elute in the same relative position to those
flanking markers. Because of this, a unique time location and window for a spectral
library entry can be set in terms of RI, thereby ensuring that metabolites don’t fall outside
the allowed window over a much longer period of time. Retention indices have
predominately been used for GC/MS methods however this approach can also have great
success for LC/MS data alignment as well. LC/MS is certainly more complex as certain
metabolites and classes of metabolites show more chromatographic shift in their RI
172 Metabolomics

markers than others, in these cases increasing the expected RI window of the library entry
in conjunction with mass and fragmentation spectrum data is sufficient for accurate
identification. The advantages over many of the widely available chromatographic
alignment tools, eg. XCMS (Smith, Want et al. 2006), as it can be used to match against a
RI locked library over long periods of time and can align data from different biological
matrices without potential distortion from structural isomers.

4.4 Identifying metabolites


Metabolite identification is essential to the biochemical and biological interpretation of the
results of metabolomic studies. Lists of integrated peak data are of little use unless a library
of spectra is available to compare peak data with to identify the metabolites represented by
those peaks. Publicly created and maintained databases do exist (Wishart, Tzur et al. 2007;
Wishart 2011). However, the utility of these databases to identify metabolites of interest
from metabolomics studies is currently limited for a number of reasons. First, due to the
significant number of different instrument types, methods, and runtimes it is a nearly
impossible task to account for every possible representative of the spectra and retention time
for a given metabolite under all of these diverse conditions. Second, metabolomics
experiments utilize a global non-targeted approach where the method is optimized to
measure as many metabolites as possible in a wide range of biological sample types (i.e.,
matrices). Certain metabolites behave differently in one matrix than in another, or
differently in the same matrix under different conditions, for example in response to an
experimental treatment versus when non-treated. Third, there may be areas of the
chromatogram with a high-degree of co-eluting metabolites. Public databases of metabolite
spectra can provide useful information in many cases, especially when no existing library
exists. However, the public information is limited and certainly not as informative or
reproducible as generating an in-house chemical reference library using the same equipment
and protocols as used to analyze the experimental samples.
While requiring a significant resource commitment, the generation of an internal library of
authentic chemical standards is a worthwhile task with significant advantages for high-
throughput metabolomics. An in-house library of authentic standards provides a clear
representation of the spectra resulting from a metabolite on the same instrument and
method used to analyze the experimental sample. A retention index for the internal library
can be calculated and set, resulting in library entries that are fixed in time. Consequently
consistent, reliable, standard spectra that do not change over time are ensured which, in
turn, facilitates automated, high confidence metabolite identification.
Software for performing spectral library matching, much like peak integration software
discussed above, is readily accessible (Scheltema, Decuypere et al. 2009). From open-source
applications to commercial packages there are numerous choices. Many software packages
use some type of forward or reverse (or both) fitting algorithm that use mass and time
components to match peaks to metabolites of similar mass and peak shape within a time
window. Due to their global, non-targeted nature, metabolomic studies are not optimized
for any metabolite in particular, so a positive metabolite identification in a metabolomics
analysis is almost never a binary decision. It is highly unlikely to simply have a positive yes
or no for a metabolite identification, instead it is more likely to have a probability score
associated with the identification. Quality control of the scoring is essential and one of the
Software Techniques for Enabling High-Throughput Analysis of Metabolomic Datasets 173

most important aspects of metabolomics analysis, especially for running studies in high
throughput.

4.5 Unnamed metabolites


A chemo-centric approach, based on a reference library, to high-throughput metabolomics is
a powerful method to identify metabolites within biological samples. If there is any
weakness to using in-house generated reference libraries it would be in the realm of
identifying the redundant ion peaks that originate from metabolites that do not exist in the
library. Methods available to identify and group these redundant ion peaks are limited
(Bowen and Northen; Dunn, Bailey et al. 2005; Wishart 2009).
The most common approach is to rely on the chromatographic elution similarity between
these redundant ions as well as looking for user defined mass relationships between the
ions that are consistent with known chemical modifications. The effectiveness of this
approach is limited in highly complex samples where metabolite co-elution is common. In
such situations, there can be multiple metabolites eluting simultaneously which
confounds identifying their respective ions based on elution. Another shortcoming of this
method is the inability to identify unique modifications or fragments that are not known
to occur.
A method that has yielded very good results for analyzing spectrometry data and fits well
within the framework of high-throughput metabolomics is the QUICS method (Dehaven,
Evans et al. 2010) This method to identify and quantify individual components in a sample,
(QUICS), enables the generation of chemical library entries from known chemical standards
and, importantly, from unknown metabolites present in experimental samples but without a
corresponding library entry. The fundamental concept of this method is that by looking at
detected ion features across an entire set of related samples, it is possible to detect subtle
spectral trends that are indicative of the presence of one or more obscure metabolites. In
other words, because of the natural biological variability of the metabolite in the study
samples, by performing an ion-correlation analysis across all samples within a given dataset
it is possible to detect ion features that are both reproducible and related to one another.
Using the cross sample correlation analysis it is then possible to add the spectral features for
that metabolite to the reference library. Then the metabolite can be detected in the future
using that library entry, even though the metabolite is unknown, i.e., without an exact
chemical identification. Importantly, this method captures any unknown metabolite because
it does not require chemical adducts and/or fragment products to be previously known or
expected. Another advantage is that statistical analysis can be used to determine whether or
not the metabolite is significant or of interest. In this way the important unnamed
metabolites can be focused on for the work of performing an actual identification which
enhances efficiency and reduces the work to identifying the most important metabolites.

5. Quality control
The ability to perform thorough quality control on identified metabolites in metabolomics
studies is extremely important. The higher the quality of data entering statistical analysis,
the higher the probability that the study will provide answers to the questions being asked.
This section will focus on three aspects of quality control – quality control samples (i.e.,
174 Metabolomics

blanks, technical replicates), software for assessing the quality of metabolite identification,
and software for assessing the original peak detection and integration. This last point may
seem out of order but for reasons to be described results in an invaluable check of the peak
quality.

5.1 Blanks – Identify the artifacts of the process


A commonly overlooked issue in biological data collection is the presence of process
artifacts. A process artifact is defined as any chemical whose presence can be attributed to
sample handling and processing and not originating from the biological sample. In all
analytical methods chemicals are inadvertently added to samples. Artifacts can include
releasing agents and softeners present in plastic sample vials and tubing, solvent
contaminants, etc. One of the easiest and most efficient means of identifying artifacts is to
run a “water blank” sample interspersed throughout the entire process alongside the true
experimental samples. In this way, the water blank will acquire all the same process-
related chemicals as the experimental samples. Consequently, identification and in silico
removal of artifacts can be accomplished by identifying those chemicals detected at
significant levels in the water blank when compared to the signal intensity in the
experimental samples. If not identified and removed, process artifacts can inadvertently
arise as false discoveries.

5.2 Technical replicates – Find the total process variation


The intrinsic reproducibility of a method is critical since it has considerable impact on
the significance and interpretation of the results. For example, if a 20% change was
detected between treatment and control samples but the analytical method had a 20%
coefficient of variation (CV) for that measurement, concerns regarding the accuracy of
the measurement would call into question the biological relevance of that change in
measurement. On the other hand, if the analytical method had a 2% CV for that same
measurement it is much more likely that the same 20% change is of “real” biological
significance. Clearly, smaller analytical variability of the method enables small, yet
meaningful, biological changes to be detected accurately and consistently. It is therefore
critical to determine the analytical reproducibility/variability of a method for every
compound/measurement.
By far the most common way to assess system stability and reproducibility is by use of
internal standards. Internal standards can be measured throughout a study to monitor
system reproducibility and stability. The drawbacks to this approach are that the number of
standards is typically small and do not represent the myriad of chemical classes typically
observed in a metabolomics analysis.
Another common approach to address method variability is by the use of technical
replicates. With this approach the same biological sample is run multiple times, e.g., in
triplicate, to determine method reproducibility. The advantage of this method over internal
standards is the ability to determine the CV of the method for each compound detected
within the matrix of the samples being analyzed. However, the disadvantage is that, while
the replicate approach is extremely effective, it is also very time-consuming and of limited
practicality in a high-throughput setting.
Software Techniques for Enabling High-Throughput Analysis of Metabolomic Datasets 175

An extremely practical and efficient approach is to run a technical replicate of a sample


composed of a small aliquot from all the samples in a study interspersed among individual
experimental samples. An aliquot of each experimental sample is pooled, then an aliquot of
the pooled sample mixture is run at regular intervals—every n number of experimental
sample injections (n to be set by operator). An advantage of this pooled sample is that it
provides CV information for all compounds detected in the study, in the matrix under
study. Another advantage is that far less instrument analysis time is required which makes
it far more practical in a high-throughput laboratory.

5.3 Quality control of automated metabolite identifications


Performing quality control (QC) for a given metabolite identification can be an exhaustive
and time-consuming task. The work to perform QC on every metabolite identification in
every sample within a metabolomics study can seem to be a nearly-impossible task.
Considering a relatively small metabolomics study of 50 samples, with an average of 800
identified metabolites per sample, there would be 40,000 spectra to review for just that one
study. Yet, as time-consuming as this process is, quality control of automated library calls is
vital for ensuring accuracy and high confidence in the data which, in turn, enables
meaningful biological interpretation of the results. A software package that can permit this
process to proceed quickly and efficiently is critical in a high-throughput setting.
Visual inspection of all the samples in a study simultaneously enables rapid metabolite
identification QC. By representing the sample data within a study as a single set in a visual
manner and creating tools that quickly allow an analyst to investigate and manually accept
or reject an automated metabolite identification, the task of performing quality control on
even extremely large datasets can be accomplished rapidly and easily. An example of a
visual data display is shown in Figure 1. In this example the panel across the top (Figure 1A)
contains a list of all of the metabolites identified by the software in the experimental samples
being analyzed. By highlighting one chemical, the structure for that compound is displayed
in an adjacent window (Figure 1B). The default visualization for viewing a highlighted
metabolite is broken down into a distinct method chart for each analytical platform method
that was used to identify that metabolite. The display also shows the multiple analytical
platforms where the metabolite was identified. In this example, the same metabolite
identified on a GC/MS platform (Figure 1C), and LC/MS negative ion platform (Figure 1D)
is shown. Within each chart, the individual sample injections, each with a unique identifier,
make up the y-axis (Figure 1E). The x-axis represents the retention index (RI) time scale.
Navigation of the interface involves scrolling down through the data table window (Figure
1A). From the interface it is also possible to review annotation regarding the highlighted
metabolite (Figure 1F), view the analytical characteristics (e.g., Mass, RI) of the metabolite as
well as toggle through RI windows containing ions characteristic of that metabolite (Figure
1G).
An example plot of data from the LC/MS negative platform is illustrated in Figure 2. In this
example the samples are initially sorted by the sample type, namely process blank, technical
replicate, or experimental sample. The dots within each method chart represent the detected
ion peaks, and each point has associated peak area, mass to charge (m/z), chromatographic
start and stop data which can be accessed by clicking on the individual dots, as shown in
Figure 3.
176 Metabolomics

Fig. 1. Graphical user interface showing the view for the proposed identification of
heptadecanoic acid. (A) Distinct list of identified metabolites for the loaded sample set. This
list includes any metabolite identified at least once in any sample with the set. It also
includes summary statistics such as averages for spectral scoring and chromatographic peak
intensities, number of times detected, and status.(B) Chemical structure for displayed
metabolite. (C) Data for the posed library identification heptadecanoic acid from the GC/MS
method. (D) Data for the posed library identification heptadecanoic acid from the LC/MS
negative ion method. (E) List of unique sample identifiers comprising the study. (F)
Comment field for storing and displaying annotations that are relevant to the currently
displayed metabolite. (G) List of other ion peaks that exist as part of the spectral library
entry. (H) List of sample sorting options including associated sample metadata; diagnosis,
group and subgroup.
Software Techniques for Enabling High-Throughput Analysis of Metabolomic Datasets 177

Fig. 2. Plot for LC/MS negative method. Individual samples in the sample set are displayed
and sorted on the y-axis. Chromatographic retention time is presented on the x-axis.
178 Metabolomics

Fig. 3. Raw MS data can be accessed for each sample via graphical user interface links. Each
“dot” represents the detected and integrated ion peak in the individual sample listed on the y-
axis. Thus, each “dot” has an associated area, height intensity, chromatographic start, stop and
apex retention time/retention index. Color and shape of “dot” are indicative of the quality of
the match to the posed library identification (see chart 1) and can be used to launch underlying
data such as raw MS data (insert). Colors of the samples listed on the y-axis also hold meaning
(see chart 1).
Software Techniques for Enabling High-Throughput Analysis of Metabolomic Datasets 179

This type of visualization permits the analyst to quickly verify the quality (i.e., QC) of the
automated peak detection and integration software. Each dot that is representative of an ion
peak can be individually removed/rejected from the proposed library identification. In this
way, extraneously detected ion peaks in the window can be visualized and individually
removed, as is the case shown in Figure 4. In this particular example there are two closely
eluting ion peaks with the same mass. One of the peaks is 2-stearoylglycerophosphocholine
(Figure 4, panel A) and the other is 1-stearoylglycerophosphocholine (Figure 4, panel B). In
both panels the correct peak must be manually approved and the incorrect peak rejected
(indicated by red dots). Stray detected ions can also be individually rejected from the
identifications. In addition, the interface permits the interrogation of the integration quality
of individual ion peaks since each dot is linked to the raw ion data as illustrated in Figure 4,
panel C and Figure 5, panel B. In this fashion any potential inaccuracy in the automated
detection and integration of individual peaks can be readily determined.
In addition to being able to curate each sample individually, the automated library
identification for an entire sample set can be rejected. An example of this is shown in Figure
5. The presence of multiple dots for each sample in the RI window (Figure 5 A) coupled
with the ability to view the underlying ion data (Figure 5B) makes it apparent that the
automated metabolite identification was based on erroneous ion peaks that resulted from
the integration of noise. As a result the automated call for the entire sample set was
manually rejected by the analyst. Accordingly, with this visualization tool the analyst can
rapidly determine the quality of the automated detection and integration and remove from
the dataset any peaks which are of questionable quality.
In addition to being able to QC the automated peak detection and integration software, an
interface such as this allows an analyst to visually inspect the quality of the library
identification in each individual sample. In the graphical plot the “dot” representing the
detected ion peak for the proposed metabolite identification is displayed in various color
and shape combinations. Each combination of color and shape within each plot is an
indicator of the quality of the automated metabolite identification, which greatly aids the
analyst in making the quality assessment rapidly. Listed in Table 1 are possible color and
shape combinations with the meaning for each. The quality assessment is based on spectral
library matching logic (Evans, Dehaven et al. 2009). This graphical display allows the
analyst to look at a proposed identification for a given metabolite made by the software and
immediately determine its quality and confidence based on spectral match scores. In this
way, the automated metabolite identifications for large datasets can be quickly evaluated by
the analyst. An example of a proposed call for a group of ions in a sample set where the
MS/MS spectral match was poor is shown in Figure 6. The low data quality is readily
apparent by the preponderance of the red colored dots in the plot.

5.4 Quality control of automated integration


GC/MS and LC/MS/MS measurements of a bio-sample usually produce millions of ions,
which are fragments and/or adducts/aggregates of the metabolites, artifacts from the
system, and potentially false ions from background noise. Ideally, the false ions from the
background noise are removed, using, for example, a Gaussian smoothing algorithm to filter
them out of the dataset. The remaining ions are then integrated across time within a mass
window to identify ion chromatographic peaks. Thousands of such ion chromatographic
180 Metabolomics

Fig. 4. Example of the visualization of closely related ion peaks. (A) Two possible ion peaks
were detected in same retention window for 2-stearoylglycerophosphocholine. In this case
the ion peak to the left is the correct peak (green and blue dots with arrow) and the peak on
the right was rejected (red). (B) The peak on the right is actually 1-
stearoylglycerophosphocholine. Therefore, the peak on the right is correct (green and blue
with arrow) while the peak on the left is rejected (red). In addition ion peaks in dashed
boxes are stray detected ion peaks not associated with the peak for 1-
stearoylglycerophosphocholine ion peaks that were rejected (red). (C) The extracted ion
chromatogram for one of the samples in the sample set for this ion shows the two peaks are
well separated and accurately integrated.
Software Techniques for Enabling High-Throughput Analysis of Metabolomic Datasets 181

Fig. 5. Example of rejected identification for entire sample set. (A) Detected ion-peaks that
result from (B) a noisy baseline as seen in one injection; the entire library identification for
this LC/MS positive method is rejected.
182 Metabolomics

Feature Color Meaning


Identified metabolite with high confidence
in the quality of the identification. These
Blue
points represent the highest level of
confidence for a metabolite identification.
Identified metabolite with some confidence
in the quality of the identification. These
Green
points require further research depending
on the context.
Identified metabolite with very low
Red confidence in the quality of the
identification.
Orange points represent a peak that was
within and near the time window but was
not called as the metabolite. There may be
cases such as for very low-level peaks that
Orange
the scoring was not adequate to make a call
but taken in context with the rest of the
dataset, the peak is indeed the metabolite in
question.
Feature Shape Meaning
Circle Regular peak.
For LC/MS a triangle represent that there
Triangle exists underlying MS/MS data to confirm a
peak’s identity.
Shapes other than a circle or triangle
represent that a user has opted to also
Other shape observe ion features that exist as part of the
library entry other than the ion feature used
for quantification.
Sample Label Color (y-axis label) Meaning
Black labels mean that there is one and only
Black one ion feature representing the metabolite
identification in the given time window.
Blue labels indicate that there is more than
one ion-feature within the time window
Blue
that may represent the metabolite
identification.
Red labels are shown when there are no
Red
possible hits for the given metabolite.
Table 1. Color/Shape combinations to demonstrate peak quality
Software Techniques for Enabling High-Throughput Analysis of Metabolomic Datasets 183

Fig. 6. An example of an entire metabolite call that was rejected because the MS/MS spectral
match was poor. (A) Red color of dots indicates that the MS/MS spectral match was of low
quality. (B) Experimental MS/MS spectrum from one injection compared to the reference
library spectrum for beta-alanyl-L-histidine (carnosine).
184 Metabolomics

peaks per sample are typically detected and integrated. Those peaks are organized into
groups within a time window, adjusted/aligned by retention index from known internal
standards to account for time drift and matched to library compounds (metabolites). For
LC/MS/MS, secondary fragment ions of the primary quantification ion can also be used to
match library compounds.
Obviously, all the profiling and analysis of metabolites in biological samples are dependent
on the accuracy and consistency of ion chromatographic peak detection and peak
integration. However, GC/MS and LC/MS/MS measurements are complicated by a
number of factors; for example, the co-elution of metabolites because of incomplete
separation, the existence of artifacts from the system, the background noise, and the
potential wide concentration ranges of metabolites in the sample. Such complexities can
affect the detection and determination of the peak start, the peak end and the peak baseline.
Incomplete separation can lead to shoulders on peaks on either the leading edge or the
trailing edge of the main peak from metabolites present at higher concentrations. When
compared to the baseline, the peak start and the peak end would be characterized by a
baseline peak or a drop peak. Because of the complexity and variance inherent in biological
samples, the same metabolite in different samples may have been automatically detected
differently in regard to peak start, peak end and peak background. For example, in some
cases, the ion chromatographic peaks for metabolites present in only trace amounts may not
be well shaped, especially when a noisy background is present, so integration of such peaks
might be quite variable from sample to sample. In other cases, the major ions for a
metabolite may appear as a small shoulder on a larger ion peak and, as a result, may not
even be detected during the automatic peak detection/integration process from sample to
sample. In still other instances, a metabolite present in high concentrations may overload the
column and distort the chromatographic profile, leading to peak splitting. For such high
concentration metabolites the automatic library match may pick only one of the two peaks
for quantification which will give an erroneously low value for the amount of the metabolite
in the sample. Clearly, each of the above examples will lead to peak detection and
integration inconsistency and inaccuracy across the samples in a sample set, which will
potentially lead to wrong conclusions and wrong decisions in later analysis.
Global metabolomics has other challenges when it comes to peak detection. Unlike targeted
metabolomics, global metabolomic profiling cannot be optimized for each metabolite that is
present within a biological sample. Chromatography methods must be broad enough to
detect as many of the metabolites in the sample as possible, regardless of chemical
characteristics. Consequently, chiral compounds cannot be resolved and structural isomers
are usually not well resolved in global metabolomics profiling. In downstream analysis for
identified metabolites, structural isomers might better be combined to represent the
metabolites, or, if one isomer is more crucial in elucidating the metabolism or biochemical
pathway, consistently picking that one form across samples would ensure analytical
consistency.
A software solution to detect and correct such inconsistencies in ion peak integration and
library matching across samples in a sample set could be developed. After the quality control
phase of the identification of the detected metabolites is completed, a deeper examination of
the consistency of peak detection and integration could be performed to ensure consistency
and accuracy. The quality control phase of automatically detected metabolites involves
Software Techniques for Enabling High-Throughput Analysis of Metabolomic Datasets 185

providing a high-quality, filtered list of identified metabolites devoid of noise and artifactual
metabolites to the end user (Figure 7). Sample set sizes range from a few samples to hundreds
and even thousands of samples. Because hundreds of metabolites can be detected and
measured in each sample, this type of quality control operates on the ‘quant’ ion peaks –those
peaks detected in the samples that are used for quantification of those metabolites.

Fig. 7. Graphical View of Peak Integration for an Identified Metabolite: Identified


metabolites (200~600) in the specified sample set (Upper Left); Quantitation peaks for
selected metabolite in the samples in the sample set (Middle Left); Type of samples and
Information about the sample peaks (Lower Left); Peak chromatograms (Upper Right);
Sample peak area (blue for original integration and red for re-integrated (Lower Right).

The chromatograms of the ion peaks representing the quantitative mass from all of the
samples in a set must be evaluated to determine if:
 the majority of the sample peaks are on the trailing edge of another peak,
 the majority of the sample peaks are on the leading edge of another peak,
 the majority are peaks that encompass two peaks in other samples, as a result of peak
splitting.
Peak integration ranges are evaluated with alignment by retention index and the statistics of
peak limits across the sample set. Accordingly, in addition to user specified manual
correction, corrections in consistency and re-integration would be suggested and presented
to the analyst for review and approval. Functionally, this type of software would give the
end user a variety of methods to both investigate the automated integration and peak calls
and to correct them as necessary. The software features must include:
186 Metabolomics

 Automatic merging of approved peaks from the sample that match to the same library
compound.
 Detection of shoulder peaks based on RI-aligned peak start or peak end distribution
across the samples.
 Manual integration
 Manual peak splitting
 Show peak chromatograms in overlay mode or tabular mode for easy review/manual
re-integration.
 Update peak integrations, peak recovery and library rematch
When an identified metabolite in a biological sample is at a sufficiently high concentration,
it can overload the column and distort the chromatographic peak. Even though it may be
out of the linear range, a consistent integration of the peak is still needed to characterize the
group of samples. Distorted peaks tend to drive the integration software to identify a less
than optimal peak to be used for quantification. In Figure 7, the peak for glucose was
incorrectly split in a handful of samples by the automated peak integrator. By examining the
consistency of the peak integration across the set of samples it is possible to easily identify
and correct this situation. As shown in the example in Figure 8, this correction would
improve the relative standard deviation from 20.1 to 7.4

6.0
4.0
2.0
0.0
6.0
4.0
2.0
0.0
6.0
4.0
2.0
0.0
6.0
4.0
2.0
Intensity/10,000,000

0.0
6.0
4.0
2.0
0.0
6.0
4.0
2.0
0.0
6.0
4.0
2.0
0.0
6.0
4.0
2.0
0.0
6.0
4.0
2.0
0.0
6.0
4.0
2.0
0.0
1850.0 1855.0 1860.0 1865.0 1870.0 1875.0 1850.0 1855.0 1860.0 1865.0 1870.0 1875.0 1850.0 1855.0 1860.0 1865.0 1870.0 1875.0 1880.0

RI

2.0

1.6
Area/100,000,000

1.2

0.8

0.4

0.0
1246500

1246512

1246524

1246534

1246545

1246557

1246580

1246592

1246604

1246616

1246628

1246640

1246652

1246676

1246688

1246700

1246712

1246724

1246736

1246748

1246770

1246778

1246786

1246793

1246800

1246808

1246816

1246828

1246832

1246836

Task ID

Fig. 8. Combining Peaks


Software Techniques for Enabling High-Throughput Analysis of Metabolomic Datasets 187

As illustrated in Figures 9 and 10, small peaks on the leading or trailing side of a larger peak
are often integrated inconsistently:
 Small shoulder peaks are detected
 Small shoulder peaks are not detected
 Small shoulder peaks are combined into the main peak
In Figure 8, the major peak on the left is identified as cysteine, whereas the shoulder on
the right side is from threonate. In one sample, the small peak from threonate was
inaccurately combined into the main peak for cysteine when it was automatically
integrated, thus inadvertently increasing the response for cysteine in that sample. After
re-integration the erroneous integration was corrected thereby restoring the correct
integration for cysteine and permitting the independent detection of threonate in the
sample as well.

Fig. 9. Examples in inconsistent shoulder peaks. Splitting of shoulder (Upper panel); Area
change after re-integration (Blue for automatic integration and red for re-evaluated
integration (Lower panel).

In Figure 10, the major peak on the right is identified as 1-docosahexaenoylglycero-


phosphocholine (1-DHGPC), whereas the shoulder on the left side is identified as 2-
docosahexaenoylglycerophosphocholine (2-DHGPC). In one sample, the peak for 2-DHGPC
was inaccurately combined into the peak for 1-DHGPC when it was automatically
integrated. In another sample, the baseline was not calculated consistently. The curves at the
lower right show the correction. After re-integration the erroneous integration was corrected
and the small peak for 2-DHGPC was recovered.
188 Metabolomics

Software that can detect inconsistencies in peak detection and integration across samples in
a sample set can ultimately improve the accuracy in the integration of peaks that have been
identified as metabolites; this in turn leads to lower CV’s and more accurate statistical
analysis which can contribute significantly to the elucidation of metabolism and metabolite
pathway.

5.0

4.5

4.0

3.5

3.0
Intensity/1,000,000

2.5

2.0

1.5

1.0

0.5

0.0
5420 5440 5460 5480 5500 5520 5540 5560 5580 5600 5620

RI

5.0
4.5
4.0
3.5
3.0
2.5
2.0
1.5
1.0
0.5
0.0
4.5
Intensity/1,000,000

4.0
3.5
3.0
2.5
2.0
1.5
1.0
0.5
0.0
4.5
4.0
3.5
3.0
2.5
2.0
1.5
1.0
0.5
0.0
5420 5460 5500 5540 5580 5420 5460 5500 5540 5580 5420 5460 5500 5540 5580 5620

RI
Software Techniques for Enabling High-Throughput Analysis of Metabolomic Datasets 189

5420 5460 5500 5540 5580 5420 5460 5500 5540 5580 5420 5460 5500 5540 5580 5620

RI

Fig. 10. Examples of inconsistent shoulder peaks

6. Conclusion
Metabolomics as a technology has demonstrated clear utility in a broad array of biological
applications. The applications are not only in demonstrating simple metabolic comparisons
between treated and control groups but in studies involving biomarker discovery, drug
development/MOA/recovery, bio-processing, agricultural applications, consumer products,
diagnostics, and so on (Sreekumar, Poisson et al. 2009; Berger, Kramer et al. 2007; Barnes, Teles
et al. 2009; Boudonck, Mitchell et al. 2009; Ma, Ellet et al. 2009; Ohta, Masutomi et al. 2009;
Watson, Roulston et al. 2009; Oliver, Guo et al.). The ability to run metabolomic studies in high-
throughput has been a challenge thus far, not so much because of the complexity or size of the
data, but because of the difficulty in generating reproducible data having low process variation
that can be quantified, is devoid of artifactual components, and provides high confidence in the
identification of metabolites. Without knowledge of the variability of the process on a
metabolite by metabolite basis, it is not possible to determine the true biological variability and
thus, cannot provide accurate answers to the questions that under investigation.
As demonstrated here, quality and the throughput of processing sample data for
metabolomics studies do not need to be mutually exclusive. By taking an intelligent
engineering approach to the data workflow, knowing when to automate a process and
developing software solutions that are streamlined for this process, the processing of sample
data for metabolomics studies can be done in significantly high volume and with high quality.

7. Acknowledgement
We gratefully acknowledge the work and contributions of all members of the Metabolon
informatics, platform, project management, statistics and management teams for their
dedicated work in building an enterprise metabolomics platform. CD, AE, HD, and KL are
employees of Metabolon.
190 Metabolomics

8. References
Barnes, V. M., R. Teles, et al. (2009). "Acceleration of purine degradation by periodontal
diseases." J Dent Res 88(9): 851-855.
Benjamini, Y. and Y. Hochberg (1995). "Controlling the false discovery rate: a practical and
powerful approach to multiple testing." Journal of the Royal Statistical Society. Series B
57: 289-300.
Berger, F. G., D. L. Kramer, et al. (2007). "Polyamine metabolism and tumorigenesis in the
Apc(Min/+) mouse." Biochem Soc Trans 35(Pt 2): 336-339.
Boudonck, K. J., M. Mitchell, et al. (2009). "Characterization of the biochemical variability of
bovine milk using metabolomics " Metabolomics 5(4): 375-386.
Bowen, B. P. and T. R. Northen "Dealing with the unknown: metabolomics and metabolite
atlases." J Am Soc Mass Spectrom 21(9): 1471-1476.
Bryan, K., L. Brennan, et al. (2008). "MetaFIND: a feature analysis tool for metabolomics
data." BMC Bioinformatics 9: 470.
Dehaven, C. D., A. M. Evans, et al. (2010). "Organization of GC/MS and LC/MS
metabolomics data into chemical libraries." J Cheminform 2(1): 9.
Dunn, W. B., N. J. Bailey, et al. (2005). "Measuring the metabolome: current analytical
technologies." Analyst 130(5): 606-625.
Evans, A. M., C. D. Dehaven, et al. (2009). "Integrated, Nontargeted Ultrahigh Performance
Liquid Chromatography/Electrospray Ionization Tandem Mass Spectrometry
Platform for the Identification and Relative Quantification of the Small-Molecule
Complement of Biological Systems." Anal Chem 81(16): 6656-6667.
Fan, T. W., A. N. Lane, et al. (2004). "The promise of metabolomics in cancer molecular
therapeutics." Curr Opin Mol Ther 6(6): 584-592.
Fields, C. (1996). "Informatics for ubiquitous sequencing." Trends Biotechnol 14(8): 286-289.
Griffin, J. L. (2006). "Understanding mouse models of disease through metabolomics." Curr
Opin Chem Biol 10(4): 309-315.
Hood, L. E., M. W. Hunkapiller, et al. (1987). "Automated DNA sequencing and analysis of
the human genome." Genomics 1(3): 201-212.
Hunkapiller, T., R. J. Kaiser, et al. (1991). "Large-scale and automated DNA sequence
determination." Science 254(5028): 59-67.
Katajamaa, M. and M. Oresic (2005). "Processing methods for differential analysis of LC/MS
profile data." BMC Bioinformatics 6: 179.
Katajamaa, M. and M. Oresic (2007). "Data processing for mass spectrometry-based
metabolomics." J Chromatogr A 1158(1-2): 318-328.
Khoo, S. H. and M. Al-Rubeai (2007). "Metabolomics as a complementary tool in cell
culture." Biotechnol Appl Biochem 47(Pt 2): 71-84.
Lawton, K. A., A. Berger, et al. (2008). "Analysis of the adult human plasma metabolome."
Pharmacogenomics 9(4): 383-397.
Lindon, J. C., E. Holmes, et al. (2007). "Metabonomics in pharmaceutical R&D." Febs J 274(5):
1140-1151.
Ma, N., J. Ellet, et al. (2009). "A single nutrient feed supports both chemically defined NS0
and CHO fed-batch processes: Improved productivity and lactate metabolism."
Biotechnol Prog 25(5): 1353-1363.
Software Techniques for Enabling High-Throughput Analysis of Metabolomic Datasets 191

Nordstrom, A., G. O'Maille, et al. (2006). "Nonlinear Data Alignment for UPLC-MS and
HPLC-MS Based Metabolomics: Quantitative Analysis of Endogenous and
Exogenous Metabolites in Human Serum." Anal Chem 78(10): 3289-3295.
Ohta, T., N. Masutomi, et al. (2009). "Untargeted metabolomic profiling as an evaluative tool
of fenofibrate-induced toxicology in Fischer 344 male rats." Toxicol Pathol 37(4): 521-
535.
Oliver, M. J., L. Guo, et al. "A sister group contrast using untargeted global metabolomic
analysis delineates the biochemical regulation underlying desiccation tolerance in
Sporobolus stapfianus." Plant Cell 23(4): 1231-1248.
Patterson, A. D., H. Li, et al. (2008). "UPLC-ESI-TOFMS-based metabolomics and gene
expression dynamics inspector self-organizing metabolomic maps as tools for
understanding the cellular response to ionizing radiation." Anal Chem 80(3): 665-
674.
Scalbert, A., L. Brennan, et al. (2009). "Mass-spectrometry-based metabolomics: limitations
and recommendations for future progress with particular focus on nutrition
research." Metabolomics 5(4): 435-458.
Scheltema, R., S. Decuypere, et al. (2009). "Simple data-reduction method for high-resolution
LC-MS data in metabolomics." Bioanalysis 1(9): 1551-1557.
Smith, C. A., E. J. Want, et al. (2006). "XCMS: processing mass spectrometry data for
metabolite profiling using nonlinear peak alignment, matching, and identification."
Anal Chem 78(3): 779-787.
Sreekumar, A., L. M. Poisson, et al. (2009). "Metabolomic profiles delineate potential role for
sarcosine in prostate cancer progression." Nature 457(7231): 910-914.
Storey, J. D. and R. Tibshirani (2003). "Statistical significance for genomewide studies." Proc
Natl Acad Sci U S A 100(16): 9440-9445.
Thielen, B., S. Heinen, et al. (2009). "mSpecs: a software tool for the administration and
editing of mass spectral libraries in the field of metabolomics." BMC Bioinformatics
10: 229.
Tolstikov, V. V. and O. Fiehn (2002). "Analysis of highly polar compounds of plant origin:
combination of hydrophilic interaction chromatography and electrospray ion trap
mass spectrometry." Anal Biochem 301(2): 298-307.
Want, E. J., A. Nordstrom, et al. (2007). "From exogenous to endogenous: the inevitable
imprint of mass spectrometry in metabolomics." J Proteome Res 6(2): 459-468.
Watson, M., A. Roulston, et al. (2009). "The small molecule GMX1778 is a potent inhibitor of
NAD+ biosynthesis: strategy for enhanced therapy in nicotinic acid
phosphoribosyltransferase 1-deficient tumors." Mol Cell Biol 29(21): 5872-5888.
Werner, E., J. F. Heilier, et al. (2008). "Mass spectrometry for the identification of the
discriminating signals from metabolomics: current status and future trends." J
Chromatogr B Analyt Technol Biomed Life Sci 871(2): 143-163.
Wilson, I. D., J. K. Nicholson, et al. (2005). "High resolution "ultra performance" liquid
chromatography coupled to oa-TOF mass spectrometry as a tool for differential
metabolic pathway profiling in functional genomic studies." J Proteome Res 4(2):
591-598.
Wishart, D. S. (2009). "Computational strategies for metabolite identification in
metabolomics." Bioanalysis 1(9): 1579-1596.
Wishart, D. S. (2011). "Advances in metabolite identification." Bioanalysis 3(15): 1769-1782.
192 Metabolomics

Wishart, D. S., D. Tzur, et al. (2007). "HMDB: the Human Metabolome Database." Nucleic
Acids Res 35(Database issue): D521-526.
Xia, J., N. Psychogios, et al. (2009). "MetaboAnalyst: a web server for metabolomic data
analysis and interpretation." Nucleic Acids Res 37(Web Server issue): W652-660.
Part 3

Metabolomics to Identify Health


Promoting Factors and New Bioactives
8

Metabolic Pathways as Targets


for Drug Screening
Wai-Nang Paul Lee1,2*, Laszlo G. Boros1,2,3 and Vay-Liang W. Go1,4
1UCLA
Center of Excellence for Pancreatic Diseases,
Los Angeles Biomedical Research Institute,
2Department of Pediatrics, Harbor-UCLA Medical Center,
3SiDMAP, LLC, Los Angeles, CA,
4Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA,

USA

1. Introduction
In the drug development process, candidate compounds are first screened for desirable
biological properties such as effects on gene expression, signal transduction, or enzyme activity.
The genetic and metabolic pathways used in the readouts are known as targets of the drug
screening process. Despite advances in molecular targeting, proteomics and metabolomics,
drug screening with molecular or metabolic targets have not produced the results that meet the
need of the pharmaceutical industry in the selection of small molecules leads/targets for clinical
testing. The relative lack of success in applying the -omics in drug screening is partly due to the
inability of the –omics to account for metabolic regulation, a property of the cellular metabolic
network. More recently, tracer-based metabolomics has been developed as an experimental
approach for the study of cellular metabolic networks. Interconversion of metabolites are
measured in terms of “extreme pathways” of the metabolic network which can be used for
drug screening purposes. In this paper, these approaches for drug screening targeting genetic
pathways (transcriptomics), biochemical pathways (metabolomics and fluxomics) and ‘extreme
pathways” (tracer-based metabolomics) are compared. The advantages and limitations of these
approaches for metabolic research and drug screening are discussed.

2. Genetic/signaling pathways as targets for drug screening


In the days of the genomic era, scientists are eager to apply the knowledge of genomics and
the advances in genetic/molecular engineering in clinical and translational research. The
general concept is that a genetic signal acts as an on-off switch in controlling metabolic
processes. However, in order to successfully apply genetic pathways (gene switches) for
drug screening, one has to establish genotype-phenotype correlation. The generally
accepted dogma of genotype-phenotype correlation is that metabolism is the final
expression of the genetic information, and peptide molecules act as signaling switches for

Corresponding author
*
196 Metabolomics

the regulation of metabolism. This popular molecular genetic approach to drug screening is
based on the assumption that the effect of drugs on metabolism and metabolic regulation is
determined by gene transcription and translation alone.
The rationale for choosing gene switches as targets for drug screening can be illustrated by the
example of the action of the tumor suppressor gene (P53) in cancer metabolism. Cancer cells
have metabolic characteristics that are distinct from normal cells in that there is an overall
increased macromolecular syntheses to sustain cell growth and proliferation. These metabolic
characteristics are generally grouped under the Warburg effect which consists of increased
anaerobic glycolysis, decreased glucose oxidation and increased glutamine utilization (1). A
representation of the model of gene switches is depicted in Figure 1. The signals that
orchestrate these metabolic changes originate from the balance between oncogenes (growth
promoting factors) that turn on signaling pathways regulating the utilization of substrates for
growth and tumor suppressor genes such as P53 that modulate energy utilization. The loss of a
cancer suppressor gene or the over-expression of an oncogene may be sufficient to generate
genetic signals to switch on or off (or modulate) metabolic pathways resulting in the cancer
cell metabolic phenotype. The interaction between molecular pathways and metabolic
pathways in cancer has recently been reviewed (1). At the molecular level, P53 regulates
transcription of genes that modulate PI3K, Akt and mTOR pathways (growth promoting
pathways) to reduce cancer growth. Excessive growth induces expression of P53 in cells
keeping cell growth and cell death in balance. Independently, P53 inhibits glucose uptake,
ribose synthesis and glycolysis thus modulating cellular metabolism. When the action of P53
is lost due to mutation, cells take up more glucose for ribose synthesis and glycolysis, the key
elements of the Warburg effect. The fact that the actions of P53 can be used to explain the
cancer metabolic phenotype suggests that any signaling pathway that interacts with P53 is a
potential target for anticancer drug screening.
The use of genetic pathways for the understanding of metabolism and drug screening has its
limitations. The interactions among signaling pathways are often based on demonstrations
using artificial overexpression or underexpression of these pathways. The real actions of these
signaling pathways in normal physiology are not exactly known. The quantitative relationship
connecting gene expression to metabolism has not been worked out. Therefore, the genetic
switch hypothesis is only one possible explanation for the expression of the cancer metabolic
phenotype. Conceptual limitations of genetic switches in the understanding of metabolisms or
the metabolic effect of drugs have been noted by D. E. Koshland Jr (2) almost half a century ago.
He pointed out that overproduction or underproduction of enzymes by molecular
manipulation may sometimes have dramatic effects on an organism and other times with only
minor effects. The overall effect of genetic manipulation on cellular metabolism cannot always
be predicted. The lack of observable effect when an enzyme concentration is changed is
analogous to the “silent” phenotypes (3) of the carrier states of many recessive diseases when
enzyme or protein concentrations of the affected genes can be substantially reduced.
Discrepancies in genotype phenotype correlation between signaling pathways and
metabolism when it occurs may be explained by our incomplete knowledge of the feedback
regulation of the signaling pathways as well as metabolic regulations of cellular metabolism.
However, the lack of genotype-phenotype correlation in many cases can be attributed to
conceptual difficulties of using genetic switches to the understand metabolism. First,
metabolic regulation is rarely an “all-or-none” type of control. According to metabolic
Metabolic Pathways as Targets for Drug Screening 197

control analysis, the regulation of metabolic pathway is distributed over many enzymes of
the biochemical reaction. Transcriptional or post-translational modification of an enzyme
potentially changes its Km and/or Vmax of the reaction. However, the change in Km or
Vmax of one enzyme may be compensated by either a change in precursor substrate
concentration or by a shift in the locus of control of the reaction to other enzymes such that
net flux remains unchanged. Secondly, the model of metabolic switches does not take into
account how the change in one metabolic pathway may impact on many other pathways
that are connected by shared substrates or co-factors and vice versa. The lack of quantitative
relationship between genotype and phenotype is the Achille’s heel of the gene switching
hypothesis† and the use of genetic pathways for drug screening.

P53 glucose P53

glucose-6-P ribose-5-P
+ NADPH

fructose-6-P

fructose-1,6-di-P

P53

lactate pyruvate

NADPH/ OAA Acetyl-CoA P53


NADP

glutamine
Malate TCA Cycle
α-KG

Fig. 1. A system of glucose metabolic pathways in a traditional format. The biochemical


pathways potentially affected by P53 as a gene-switch are indicated.

†Gene expression can be quantitatively determined using RTPCR method. Results are reported in folds

of change. Even though there may be a correlation between the fold of change and the observed
metabolic effect, the correlation is not a quantitative one.
198 Metabolomics

3. Biochemical pathways as targets for drug screening


In the past decades, metabolite profiling (metabolomics) and fluxomics have been
developed to fill in the knowledge gaps of gene regulations of metabolism. Thus genomics,
transcriptomics, metabolomics and fluxomics are the popular –omics of systems biology‡.
The advances in mass spectrometry and nuclear magnetic resonance spectroscopy have
enabled the new industry of metabolomics. These technologies provide quantitative and
qualitative analyses of organic compounds in biological fluids and specimens. Quantitation
of metabolites at different time points is the basis of fluxomics. Research in metabolite
profiling and fluxomics is based on our understanding of metabolic control analysis (MCA).
MCA provides quantitative measures of degree of influence of a change in enzyme kinetics
or a change in substrate concentration can affect the consumption or production of a
metabolite in terms of metabolic control coefficients (5, 6). Thus, measurements of substrate
concentrations by metabolite profiling and flux analyses allow detail information regarding
metabolic changes when the system is perturbed by drug treatment. In metabolomics and
fluxomics, traditional biochemical pathways can be considered as targets for drug candidate
screening. Such screening has the limitation in that the effect on the metabolic system as a
whole is not evaluated in the screening process. Technically, current metabolomics
technologies do not permit characterization of substrate concentrations at the subcellular
level and reactions that are compartmentalized cannot be properly evaluated. The sampling
processes usually do not separate the contribution from background environment such as
the culture medium or neighboring cells to the metabolic processes of the cell. For these
reasons, metabolomics has not been successfully used as targets for drug screening.
Measurements of flux (fluxomics) depend on the use of isotopes (7). Since 13C labeled
isotopes can be distributed widely among many metabolites, and not all of these
metabolites can be measured in the same analytical method, there are always fewer data
than needed to give precise quantification of flux. Nevertheless, this approach has its
appeal in that once a mathematical model is constructed, literature values can be fitted
into the model to give insight into possible changes in the system, and whether the model
is robust or not can be tested (8). Such an approach was used by Selivanov et al. to model
of the pathways of pentose phosphate cycle. The interconnection of these pathways is
shown in Figure 2 (9). After incubating cells with [1, 2 13C2]-glucose, ribose was found to
be labeled in many carbon positions. Using mass isotopomer distribution in ribose and
known sugar phosphate concentrations and Km values of enzymes from the literature,
these authors were able to simulate the fluxes of the pentose phosphate pathways. They
were able to identify three reactions among other transketolase mediated reactions that
were significantly inhibited when cells were treated with oxythiamine, a tranketolase
inhibitor. These are xylulose-5p to glyceraldehyde-3-P, sedoheptulose-7-p to ribulose-5-P
and xyluose-5-P to sedoheptulose-7-P (reactions 14, 15 and 13 in figure 2). The differences
in response among tranketolase enzymes inhibited by oxythiamine are the consequence of
stoichiometric constraints.

‡Systems biology as commonly defined is the enumeration of a collection of biologically related objects
(genomics, proteomics and metabolomics) or characteristics (transcriptomics and fluxomics) within the
boundary of a cell. However, in actuality the context of a cellular boundary i.e. how these objects or
characteristics separate the cell from its environment is often absent in the definition of these systems (4).
Metabolic Pathways as Targets for Drug Screening 199

Fig. 2. A model of pentose phosphate cycle used for carbon tracing and flux analysis. Using
experimentally determined isotopomer distribution in ribose and lactate, the fluxes of the
numbered reactions can be calculated (from reference 9 with permission).

The fluxomic approach targeting traditional biochemical reactions provides more specific
information regarding the metabolic system than metabolite profiling (metabolomics). The
use of fluxomics allows the simultaneous assessment of the effect of a drug on multiple
metabolic pathways and permits a better understanding of metabolism than the gene-
targeting approach. However, in order to take into account futile cycling or stoichiometric
constraints, stable isotope tracing (carbon tracing) is required as illustrated in the above
example. Even though it is possible to construct a complex model for mammalian metabolic
networks to take into account of futile cycles and stoichiometric constraints, such a model
requires a very large data set and extensive programming. In the best case scenario, there is
never sufficient data for solving all the parameters of the system and the results are model
dependent and are difficult to verify for practical reasons (2). Nonetheless, the fluxomics
approach definitely provides better correlation with phenotype than the gene switch
targeting approach.

4. “Extreme pathways” and metabolic network


The “extreme pathways” of a metabolic network can also be used as targets for drug
screening. “Extreme pathways” are elements of the well known constraint-based modeling
(8) which has been applied to the study of cellular homeostasis. (The definition of “extreme
pathway” is given in the next section.) Living organisms (cells) are metabolic systems
(networks) continuously exchanging energy substrates with their environments to maintain
the biological systems in a homeostatic state. The main metabolic function of a cell is to
200 Metabolomics

utilize substrates from its environment to produce energy and building material for the
synthesis of macromolecules. Excess intermediates are returned to the surrounding
environment to maintain a relatively constant internal environment. The boundaries of
metabolic activities represented by “extreme pathways” within which the cell functions
define the homeostatic state (10, 11). These boundaries are the result of constraints by the
stoichiometry of competing reactions, synchronization of shared pathways and/or
intermediates, and balance of energy production and utilization.
The role of “extreme pathways” in the maintenance of homeostasis can be illustrated by the
example of glucose metabolism via the TCA cycle. Pyruvate from glycolysis is metabolized
via pyruvate carboxylation leading to the conservation of 3-carbon species or pyruvate
decarboxylation leading to production of 2-carbon species (via acetyl-CoA) and energy
production (beta-oxidation and tricarboxylic acid (TCA) cycle) (16). These two processes are
concurrent in cells and the activity of one pathway constrains the activity of the other. For a
given homeostatic state, the observed utilization of pyruvate via these pathways is the
optimal§ pyruvate utilization and can be represented by a vector in the pyruvate phenotypic
phase plane. The operation of the TCA cycle is an example of metabolic constraint due to
synchronization of shared pathways or intermediates. A full turn of the TCA cycle oxidizes a
mole of acetate into two moles of carbon dioxide with production of reducing equivalents
and/or high energy phosphates. At the same time each of the TCA cycle intermediate may
have its respective substrate cycle such as the malate cycle and the citrate lyase cycle. These
individual substrate cycles perform separate metabolic functions in conveying reducing
equivalents (malate shuttle) and acetyl-CoA (citrate lyase cycle) from the mitochondria to the
cytosol. The operations of these cycles are usually synchronized for efficiency. When there is
a lack of synchrony of these cycle, abnormal substrate and energy balance can result and a loss
of homeostasis in the cell occurs. The imbalance of energy metabolism in the mitochondria due
to imbalance of substrate cycles is a frequent cause for reactive oxygen species generation and
apoptosis. Changes in these boundaries consisting of “extreme pathways” are sensitive to
metabolic or therapeutic perturbations and are excellent markers of therapeutic effects.
The differnces between a metabolic network and a traditional biochemical reaction model
can best be shown by representing a metabolic network as an engineering system. The
working of such a system is illustrated in figure 3 in which pathways shown in Figure 2 are
represented as belts and wheels connecting glycolytic/gluconeogenic substrates to those of
the pentose cycle. The enzymes that drive the belts are indicated and the role of energy
production and utilization are included. Figure 2 is model of pentose cycle intermediates
linked by enzymatic reaction. The fluxes of these reactions can be modeled mathematically
using a set of ordinary differential equations. Figure 3 shows the production and
consumption of different classes of compounds connected to the production and
consumption of ATP and reducing equivalents. These models are conceptually different.
The input-output model of cellular homeostasis of tracer-based metabolomics can account
for stoichiometric constraints and synchronization of substrate cycles thus overcoming
limitations of the previous approaches in metabolic studies.

§Optimality is sometimes thought of as a teleological concept. The optimal metabolic function of a cell

is not for its purpose to survive, but is defined by the internal organization of the metabolic network.
Metabolic Pathways as Targets for Drug Screening 201

Fig. 3. An engineering model of the system of reactions depicted in Figure 1 and Figure 2.
The relationship among the different substrate pools is represented by different circles.
Stoichiometric relationships are provided by the mass balance equations. The productions of
these metabolites from one another are indicated by respective drive belts. Energy substrate
consumption and production is also included in the model. The metabolic network and its
function is shown as a factory production model with sources and sinks of the raw materials
and products.

The basic concept and tracer methodology of tracer-based metabolomics have been
reviewed (4, 10-12). A key feature that distinguishes tracer-based metabolomics from
metabolite profiling (metabolomics) and fluxomics is the inclusion of a system boundary
that permits input-output analysis and a balance of flux** model in which substrate input is
link to its output (products) by “extreme pathways” (12, 13, 14). “Extreme pathways” are
pathways that elements (carbon, oxygen and nitrogen) from compounds (precursors)
introduced into the system travel over to the final products. The basic elements of “extreme
pathways” form the axes of a high dimension phenotypic space, any two of these axes forms
a phenotypic phase plane and the line of optimality which is a vector within the space (or a
plane) representing the metabolic phenotype. The relationship among any three “extreme

A balance of flux analysis requires a steady state or quasi-steady state assumption. For most cellular
**

processes involving cell growth and division, these processes are slow relative to the experimental
study period and quasi-steady state of metabolic reactions may be safely assumed. However, in
biological processes that are fast such as muscle contraction, or nerve conduction the balance of flux
model cannot be applied and a dynamic model is required.
202 Metabolomics

pathways” can be described by isoclines (15). Therefore, tracer-based metabolomics offers a


graphical representation of a quantitative metabolic phenotype not available by gene-
switching model or fluxomic model.
Instead of measuring substrate fluxes over specific biochemical reactions, the
experimental model focuses on fluxes over “extreme-pathways” which are pathways
linking the precursor substrates to the specific products. There may be many “extreme
pathways” for the formation of a product depending on how many interconnecting
reactions between precursor and end-product. In the synthesis of glutamic acid from
glucose, there are at least two “extreme pathways” namely the pyruvate carboxylase and
pyruvate dehydrogenase reactions (4). Pentose is synthesized from glucose at least
through two “extreme pathways” either by oxidative (G6PDH) or non-oxidative (TK/TA)
pathways (10).

5. Measuring “extreme pathways” – Carbon tracing in tracer-based


metabolomics
A unique feature of tracer-based metabolomics as the name implies is the application of
stable isotope labeled metabolites and mass spectrometry or magnetic resonance (NMR)
spectroscopy. The 13C or 2H label from the labeled substrate is distributed into metabolic
intermediates in specific positions according to the “extreme pathways”. Tables 1a and 1b
show some of the examples of labeling in amino acids, glycogen, ribose and lactate from
uniformly labeled glucose [U13C6]-glucose (carbon tracing from glucose) (16-24). The tables
show the potential mass isotopomers that can be generated, the positions that are labeled in
the products, and the corresponding glucose carbon that the 13C originates. For example,
three mass isotopomers (M3, M2 and M1 can be found in alanine or lactate from an
experiment with [U13C6]-glucose. M3 of alanine comes from glycolysis of glucose. The 13C in
carbon 1 of alanine comes from carbon 3 of glucose (G3). There are two M2 isotopomers
with 13C on either C3 and C2 or C2 and C1 of alanine. The sources of 13C’s are from glucose
carbon 2 (G2) and carbon 1 (G1). X represents a 12C carbon originated from exchange at the
level of the TCA cycle. The mass isotopomers and positions with 13C label in these glucose
metabolic intermediates are indicated. These tables can be used as a guide to design tracer
studies or interpret results from such studies. The mass and position isotopomers in these
metabolites represent individual “extreme pathways” from glucose carbon to the respective
products. It should be noted that some of the isotopomers are products of the same
“extreme pathways” thus providing redundancy in the information on the “extreme
pathways”. In the example of labeled glucose forming labeled amino acids (Table 1a), we
can gain insight into the simultaneous reactions of pyruvate carboxylation, pyruvate
dehydrogenase, malate cycle, gluconeogenic cycle relative to TCA cycle flux. When the
distribution of isotopomers is determined using mass spectrometry or NMR, we can use
these isotopomer ratios to construct phenotypic phase planes (11, 15). Such a database of
mass isotopomers can easily be managed with computational algorithm (subroutines) which
can compare distances between individual phenotypes on different phenotypic phase
planes. Phenotypic differences can also be quantitatively compared using isocline analysis.
Thus, tracer-based metabolomics is a quantitative experimental approach to the study of
metabolism and metabolic regulation.
Metabolic Pathways as Targets for Drug Screening 203

Isoto- Alanine Serine Glycine Asparate Glutamate


Pathway
pomer C3-C2-C1 C3-C2-C1 C2-C1 C4-C3-C2-C1 C5-C4-C3-C2-C1

G2-G1-G1-G2-
M5 --- --- --- ---
G3@

G2-G1-G1- G2-G1-G1-G2-X;
M4 --- --- ---
G2@ G2-G1-G1-G1-X

M3 G1-G2-G3 G1-G2-G3 --- X-G1-G2-G3


Glycolysis
Plus G2-G1-X G2-G1-X X-G1-G2-X G2-G1-X-X-X
TCA Cycle M2 G2-G1
X-G2-G1 G2-G1-X-X

G1-X-X G1-X-X-X, X-X-G1-X-X


M1 X-G1-X X-X-G1 --- X-G1-X-X X-X-X-G1-X
X-X-G1 X-X-G1-X X-X-X-X-G1

M3 --- G1-G2-G3 --- --- ---


1-Carbon
M2 --- G2-G3 G2-G3 --- ---
Metabolism
M1 --- G3 G3 --- ---

The orientations of the amino acid molecules are shown in the top row. Mass isotopomers are
designated as M1 to M5 indicating the number of 13C per molecule of the amino acid. The
corresponding position of glucose carbon within the amino acid is designated as G1 to G3. The glucose
molecule is symmetrical around C3-C4. In the table, G1-G2-G3 is the same as G6-G5-G4, if these
positions are labeled equally. X represents 12C from exchange within the TCA cycle.
@ When glucose enrichment is high, there is a likelihood of labeled OAA condensing with labeled acetyl-

CoA resulting in M5 –ketoglutarate and subsequently M5 glutamate and M4 aspartate.

Table 1a. Examples of Position and Mass Isotopomer Distribution in Gluconeogenic Amino
Acids from [U13C6]-glucose (16-24)
204 Metabolomics

Glycogen Ribose Lactate


Pathway Isotopomer
C6-C5-C4-C3-C2-C1 C5-C4-C3-C2-C1 C3-C2-C1
Hexose-P cycles M6 G6-G5-G4-G3-G2-G1 --- ---
G6-G5-G4-X-X-X G1-G2-G3
M3
Glycolysis X-X-X-G3-G2-G1
G6-G5-X-X-X-X G2-G1-X
M2
Glycolysis X-X-X-G3-G2-X X-G2-G1
Plus G1-X-X X-X-X G1-X-X
TCA Cycle M1 X-G1-X-X-X-X X-G1-X
X-X-G1-X-X-X X-X-G1

Oxidative G6-G5-G4-G3-
M5
G2
Non-oxidative Glycogen glucose are
M4 labeled as in G3-G2-X-G3-G2
glycolysis/
M3 G3-G2-X-X-G3
Oxidative gluconeogenesis
shown above X-X-X-G3-G2
plus M2
G3-G2-X-X-X
Non-oxidative
M1 G3
The orientations of the glycogen, ribose and lactate molecules are shown in the top row. Mass
isotopomers are designated as M1 to M6 indicating the number of 13C per molecule of the amino acid.
The corresponding position of glucose carbon within the glycogen, ribose and lactate is designated as
G1 to G6. The glucose molecule is symmetrical around C3-C4. In the table, G1-G2-G3 is the same as G6-
G5-G4, if these positions are labeled equally. X represents 12C from exchange within the TCA cycle.
Table 1b. Examples of Position and Mass Isotopomer Distribution in Glycogen, Ribose and
Lactate from [U13C6]-glucose (16-24)

6. “Extreme pathways” as targets for drug screening


The application of phenotypic phase plane (PPP) analysis of balance of flux data from tracer-
based metabolomics is a graphical way of presenting experimental data that is unique to
tracer-based metabolomics. Since metabolic phenotype of a cell is characterized by the
pattern of its utilization of substrates, the phenotype of a cell is represented by the
input/output characteristics which can be measured as fluxes through the “extreme
pathways”. Any two of these “extreme pathways” can form a phenotypic phase plane. The
metabolic phenotype of a cell is given by a vector in the plane and the vector divides the
plane into two regions representing regions of relative excess and relative deficiency of
substrate utilization (4). The use of phenotypic phase plane analysis together with isocline
analysis allows quantitative comparison of different treatment effects. An example of the
use of isocline analysis is illustrated by the study of effects of fructose and glutamine on the
glycolytic/gluconeogenic pathways (11).
Other applications of phenotypic phase plane analysis to investigate metabolic mechanisms
of a therapeutic intervention are illustrated in Figure 4. Panel (i) of the figure shows the
metabolic response of a cell to two therapeutic interventions (e.g. drugs, receptor inhibitor,
Metabolic Pathways as Targets for Drug Screening 205

Fig. 4. Examples of phenotypic phase plane analysis showing the quantitative relationship
among phenotypes by isocline analysis. Panel (i) shows effects of two drugs (A and B) with
different mechanisms on metabolic pathways X and Y. N represents the normal phenotype.
Panel (ii) shows the effect of A is orthogonal to the X-Y plane. Panel (iii) shows dose
dependent effect of A. Panel (iv) shows non-linear response to two different doses of A.

or siRNA) A and B. These treatments result in changes in phenotypes (decrease in


production of Z) accompanied by different metabolic compensations in substrate utilization.
Treatment B results in a decreased utilization of substrate X (or its metabolic pathways)
which is compensated by a slight increase in the utilization of substrate Y. Whereas
treatment A results in the increase utilization of X and decreased utilization of Y as
compared to control (N). If the mechanism of action of treatment B is known (such as
inhibition of a specific kinase), one can conclude that treatment A must act on a different set
of metabolic and/or signaling pathways. Such an approach will allow an iterative approach
to the discovery of new treatment or new pathways. Additional examples of using
phenotypic phase plane to understand phenotype changes of fibroblasts from a patient with
thiamine responsive megaloblast anemia (TRMA) and pancreatic cancer cell (MIA) and
normal fibroblasts using PPP have been provided by Lee and Go in their review (15). When
TRMA cells were treated with high doses of thiamine, the phenotype approached that of the
normal fibroblasts. On the other hand, MIA cells had a high pentose synthesis phenotype
which was corrected when they were treated with oxythiamine. (15) We have recently used
phenotypic phase plane analysis to show that inhibition of histone acetylation by genetic
206 Metabolomics

intervention or by chemical inhibitor of the reaction had similar metabolic phenotypes (25).
Panel (ii) shows result of treatment A which is orthogonal to the phenotypic phase plane of
X and Y. This means that treatment A affects a different part of the metabolic system which
is not linked to the utilization of substrates X and Y. Metformin and rosiglitazone, two
antihyperglycemic drugs, have been shown to alter de novo lipogenesis. While inhibition of
de novo lipogenesis by metformin is in the plane of ribose metabolism, meaning changes in
pentose cycle metabolism is related to the decrease in lipogenesis. On the other hand, the
increase in fatty acid synthesis by rosiglitazone is orthogonal to the ribose phenotypic phase
plane suggesting very different mechanism of actions by these two drugs (26). The ability to
detect orthogonal phenotypic phase plane is important because there are potentially many
of these orthogonal phenotypic phase planes which can be discovered using tracer-based
metabolomics, and each of the orthogonal pair would suggest different mechanism of action
by different drugs. The finding of orthogonal planes is one of the unique capability of the
metabolomics approach in generating mechanistic hypothesis. Panel (iii) shows the
proportional response to inhibitor of substrate X where all of the isoclines are parallel to
each other. An example of this type of response is provided by our study on the response of
a methotrexate resistant colon cancer cell line (HT29) to the effect of DHEAS, oxythiamine
and methotrexate treatment alone and in combination (27). Panel (iv) shows response to
two inhibitors of substrate X with non-linear compensation of substrate Y. The application
of PPP analysis has allowed a far better understanding of metabolic adaptation in cellular
homeostasis using tracer-based metabolomics. Using PPP and isocline analysis, we can
directly exploit the large dataset accumulated from tracer-based metabolomics studies for
target discovery and lead identification in pharmaceutical industry.

7. Concluding comments
Study of metabolism in the post-genomic era differs from the traditional biochemistry in
that the study is focused on the function of the system of biochemical reactions in a cell (or
the cellular metabolic network) and its regulation. Metabolic function of a living organism
(cell) is what mediates the the genetic potential of a cell and its interaction with its
environment to maintain homeostasis. The maintenance of homeostasis by the cellular
metabolic network in a living organism is the basis of normal physiology and histology (28).
When the metabolic environment of a living organism or a cell is altered such as in diabetes
or metabolic diseases, maladaptation or the lack of homeostasis in the living organism is the
underlying cause for pathophysiology and histopathology (29).
Metabolic phenotype of a cell is the result of genetic and environmental interaction.
Understanding metabolic phenotyping changes is important to our understanding of how cells
maintain homeostasis by metabolic regulation. We have reviewed three approaches that are
used in such investigations based on three different models. Of these different approaches, the
gene-switch approach is the most extensively used in the pharmaceutical industry. In the gene-
switch model, metabolic regulation begins with the interaction between genes and signaling
pathways which eventually impact on biochemical reactions known as down-stream effects
(30). This model ignores the fact that many of these signaling pathways or transcriptional
factors are altered through post-translational events such as phosphorylation, acetylation,
glycosylation and methylation. Since all these post-translational modifications are basic
biochemical reactions, they are all subject to the stoichiometric and energy substrate constraints.
Metabolic Pathways as Targets for Drug Screening 207

Once the downstream events are initiated, the interconversion of metabolic intermediates is
subject to all the constraints as described in preceding paragraphs. We have previously shown
that altered metabolic pathways can be the initiating events in gene transcription and post-
translational modification of signaling pathways and enzymes (31, 32). Therefore, gene-switch
model is an incomplete model to understand metabolic regulation. Because of these conceptual
problems, application of gene-switch approach has had disappointing results in identifying
drug candidates or targets and appalling failures in clinical trial due to unexpected toxicity or
lack of efficacy. Of the two remaining approaches, tracer-based metabolomics is a practical
experimental approach that does not require complicated mathematical modeling.
Furthermore, the results can be graphically presented and the quantitative difference of
metabolic phenotypes can be compared. Such features make the tracer-based metabolomics a
powerful approach for drug screening in pharmaceutical research. Since the model does not
assume any signaling pathways, it is most suitable for studies of nutriceuticals such as
phytochemicals (33) or for screening of compounds in a chemical library that have no known
molecular targets (34). It is also applicable to investigate the metabolic effect of drug
combinations, in which the interaction of drugs can be studied. Most important of all, tracer-
based metabolomics approach provides the understanding of cellular homeostasis and its
changes under the influence of nutrient conditions or pharmaceuticals.
Since our first publication on metabolic profiling (35), progress in tracer-based
metabolomics has been slow because there are few investigators who are trained in tracer
technology. The current tracer model mainly addresses the area of glucose metabolic
pathways. Methods for the investigation of other metabolic systems that may be distantly
connected to glucose metabolism (orthogonal systems) are not represented. These systems
include the systems of glutamine metabolism which connects glucose metabolism to nucleic
acid synthesis; arginine metabolism which is part of the urea cycle and nitric oxide synthesis
system; and the methyl donor pathways which are important in nucleic acid synthesis and
choline synthesis. The complete development of tracer-based metabolomics is probably a
decade away provided the development has the attention and adequate funding to complete
the tasks to cover the metabolic pathways of the whole cellular metabolic network.

8. Acknowledgement
This work is supported by the Biomedical Mass Spectrometry Laboratory of the GCRC at
the Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center (PHS M01-
RR00425; UCLA; CTSI 1UL1-RR033176) and the Metabolomics Core Laboratory of the
UCLA Center of Excellence in Pancreatic Diseases (P01 AT003960).

9. References
[1] Levine AJ, Puzio-Kuter AM. The Control of the Metabolic Switch in Cancers by
Oncogenes and Tumor Suppressor Genes. Science. 2010 Dec 3;330(6009):1340-4.
[2] Koshland Jr. DE Control of enzyme activity and metabolic pathways. pp1-8, in
“Metabolic Regulation” (eds. R.S. Ochs, R.W Hanson and J Hall), Elsevier Science
Publisher, Amsterdam (1985).
208 Metabolomics

[3] Raamsdonk LM, Teusink B, Broadhurst D, Zhang N, Hayes A, Walsh MC, Berden JA,
Brindle KM, Kell DB, Rowland JJ, Westerhoff HV, van Dam K, Oliver SG. A
Functional Genomics Strategy That Uses Metabolome Data to Reveal The
Phenotype of Silent Mutations. Nat. Biotechnol 2001; 19:45–50.
[4] Cascante, M., Comin, B., Boren, J., Raïs, B., Centelles, J.J., Puigjaner, J., Lee, W.-N. P.,
Boros, L.G. "Application of Metabolic Control Analysis to the Design of a New
Strategy for Cancer Therapy", pp. 173-180 in "Technological and Medical
Implications of Metabolic Control Analysis" (ed. A. Cornish-Bowden and M. L.
Cárdenas), Kluwer Academic Publishers, Dordrecht (2000)
[5] Cascante M, Boros LG, Comin-Anduix B, de Atauri P, Centelles JJ, Lee PW.Metabolic
control analysis in drug discovery and disease. Nat Biotechnol. 2002 Mar;20(3):243-
9.
[6] Lee WN. Characterizing Metabolic Phenotype Using Tracer-Based Metabolomics.
Metabolomics 2006, 2:31-39.
[7] Zamboni N, Fendt SM, Rühl M, Sauer U. (13)C-based metabolic flux analysis. Nat
Protoc. 2009;4(6):878-92. Epub 2009 May 21.
[8] Selivanov VA, Puigjaner J, Sillero A, Centelles JJ, Ramos-Montoya A, Lee PW, Cascante
M. An Optimized Algorithm for Flux Estimation From Isotopomer Distribution in
Glucose Metabolites. Bioinformatics 2004; 20:3387-3397.
[9] Selivanov VA, Meshalkina LE, Solovjeva ON, Kuchel PW, Ramos-Montoya A, Kochetov
GA, Lee PW, Cascante M. Rapid Simulation and Analysis of Isotopomer
Distributions Using Constraints Based on Enzyme Mechanisms: An Example From
HT29 Cancer Cells. Bioinformatics 2005; 21:3558-3564.
[10] Lee WN, Go VL. Nutrient-Gene Interaction: Tracer-Based Metabolomics. J Nutr 2005;
135:3027S-3032S.
[11] Lee, WN, Wahjudi PN, Xu, J, Go VLW. Tracer-based Metabolomics: Concepts and
Practices. Clin Biochem. 2010 Nov;43(16-17):1269-77.
[12] Maguire G, Boros LG and Lee P. Development of Tracer-Based Metabolomics and its
Implications for the Pharmaceutical Industry. Int J Pharm Med 2007; 21 (3): 217-
224.
[13] Schilling CH, Palsson BO. The Underlying Pathway Structure of Biochemical Reaction
Networks. Proc Natl Acad Sci 1998; 95:4193-4198.
[14] Schilling CH, Letscher D, Palsson BO. Theory for the Systemic Definition of Metabolic
Pathways and Their Use in Interpreting Metabolic Function From a Pathway-
Oriented Perspective. J Theor Biol 2000; 203:229-248.
[15] Edwards JS, Ramakrishna R, Palsson BO. Characterizing the Metabolic Phenotype: a
Phenotype Phase Plane Analysis. Biotechnol Bioeng. 2002; 77:27-36.
[16] Katz J., Lee WP. The Application of Mass Isotopomer Analysis in The Determination of
Pathways of Glycogen Synthesis. Am. J. Physiol 1991; 259:E757-E762.
[17] Lee WNP, Boros LG, Puigjaner J, Bassilian S, Lim S, Cascante M. Investigation of The
Pentose Cycle Using [1, 2-13C2]-Glucose and Mass Isotopomer Analysis:
Estimation of Transketolase and Transaldolase Activities. Am. J. Physiol 1998; 274:
E843-E851.
Metabolic Pathways as Targets for Drug Screening 209

[18] Katz J, Lee WNP, Wals PA, Bergner EA. Studies of Glycogen Synthesis and the Krebs
Cycle by Mass Isotopomer Analysis with U-13C-Glucose in Rats. J. Biol. Chem
1989; 264:12994-13001.
[19] Katz J, Wals PA, Lee WP. Determination of Pathways of Glycogen Synthesis and
Dilution of the 3-Carbon Pool with [U-13C6]-Glucose. Proc. Natl. Acad. Sci 1991;
88:2103-2107.
[20] Katz J, Wals PA, Lee WP. Isotopomer Studies of Gluconeogenesis and the Krebs Cycle
with 13C Labeled Lactate. J. Biol. Chem 1993; 268:25511-25521.
[21] Lee WNP. Appendix: Analysis of Tricarboxylic Acid Cycle Using Mass Isotopomer
Ratios. J. Biol. Chem 1993; 268:25522-25526.
[22] Lee WP, Edmond J, Bassilian S, Morrow J. Mass Isotopomer Study of Glutamine
Oxidation and Synthesis in Primary Culture of Astrocytes. Develop. Neurosci 1996;
18:469-477.
[23] Fu TF, Rife JP, Schirch V. The Role of Serine Hydroxymethyltransferase Isozymes in
One-Carbon Metabolism in MCF-7 Cells as Determined by (13)C NMR. Arch
Biochem Biophys 2001; 393:42-50.
[24] Solà A, Maaheimo H, Ylönen K, Ferrer P, Szyperski T. Amino Acid Biosynthesis and
Metabolic Flux Profiling of Pichia Pastoris. Eur J Biochem 2004; 271:2462-2470.
[25] Alcarraz-Vizán G, Boren J, Lee WN, Cascante M. Histone deacetylase inhibition results
in a common metabolic profile associated with HT29 differentiation. Metabolomics.
2010:229-237.
[26] Personal unpublished observation.
[27] Ramos-Montoya A, Lee WN, Bassilian S, Lim S, Trebukhina RV, Kazhyna MV, Ciudad
CJ, Noe V, Centelles JJ, Cascante M. Pentose phosphate cycle oxidative and
nonoxidative balance: A new vulnerable target for overcoming drug resistance in
cancer. Int J Cancer. 2006:119(12):2733-41.
[28] Boros, LG, Lee WNP and VLW. Go. A Metabolic Hypothesis of Cell Growth and Death
in Pancreatic Cancer. Pancreas 2002: 24:26-33.
[29] Boros LG, Steinkamp MP, Fleming JC, Lee WNP, EJ Neufeld. Defective RNA Ribose
Synthesis in Thiamine-Responsive Megaloblastic Anemia (TRMA): Mechanism for
the Syndrome. Blood 2003; 102:3556-3561.
[30] Zanuy M, Ramos-Montoya A,• Oscar Villacanas O, Canela N, Miranda A, Aguilar E,
Agell N, Bachs O, Rubio-Martinez J, Pujol MD, Lee WNP, Marin S, Cascante M.
Cyclin-dependent kinases 4 and 6 control tumor progression and direct glucose
oxidation in the pentose cycle. Metabolomics 2011 DOI 10.1007/s11306-011-0328
[31] Zhang H, Cao R, Lee WN, Deng C, Zhao Y, Lappe J, Recker R, Yen Y, Wang Q, Tsai MY,
Go VL, Xiao GG. Inhibition of protein phosphorylation in MIA pancreatic cancer
cells: confluence of metabolic and signaling pathways. J Proteome Res. 2010 Feb
5;9(2):980-9. PMID: 20035555; PMC2836017
[32] Ma D, Wang J, Zhao Y, Lee WNP, Xiao J, Go VL, Wang Q, Recker R, Xiao GG Inhibition
of glycogen phosphorylation induces changes in cellular proteome and signaling
pathways in MIA pancreatic cancer cells Pancreas 2011
D01:10.1097/mpaOBO13E318236F022
210 Metabolomics

[33] Harris DM, Li L, Chen M, Lagunero FT, VLW Go, Boros LG. Diverse mechanisms of
growth inhibition by luteolin, resveratrol, and quercetin in MIA PaCa-2 cells: a
comparative glucose tracer study with the fatty acid synthase inhibitor C75
Metabolomics 2011 DOI 10.1007/s11306-011-0300-9
[34] Harrigan GG, Brackett DJ, Boros LG. Medicinal chemistry, metabolic profiling and drug
target discovery: a role for metabolic profiling in reverse pharmacology and
chemical genetics. Mini Rev Med Chem. 2005 Jan;5(1):13-20.
[35] Boros LG, Cascante M, Go VLW, Heber D, Hidvégi M, Lee WNP. Metabolic Profiling of
Cell Growth and Death in Cancer: Applications In Drug Discovery. Drug Discovery
Today 2002; 7:366-374.
Part 4

Metabolomics in Plant Research


9

New Opportunities in Metabolomics and


Biochemical Phenotyping for Plant
Systems Biology
Gibon Yves1,2, Rolin Dominique2,3, Deborde Catherine1,2,
Bernillon Stéphane1,2 and Moing Annick1,2
1INRA, UMR1332 Fruit Biology and Pathology,
Centre INRA de Bordeaux, Villenave d'Ornon
2Metabolome Facility of Bordeaux Functional Genomics Centre,

Centre INRA de Bordeaux, Villenave d'Ornon


3Université de Bordeaux, UMR1332 Fruit Biology and Pathology,

Centre INRA de Bordeaux, Villenave d'Ornon


France

1. Introduction
Today’s unsustainable use of fossil fuel reserves or green fuel is predicted to destabilize the
global climate and lead to reduced food security. The key challenge for the coming decades
are to meet local needs for food, in terms of both quantity and quality, while conserving
natural resources and biodiversity (Ruane & Sonnino, 2011) and to develop a supply
industry based on renewable plant-derived products. Indeed agricultural crops can be
viewed as a source of or starting point for a plant based economy, potential input to a bio
refinery in which all parts of the plant are processed and used to yield (i) food, both
traditional and with enhanced nutritional safety, stability and processability; (ii) industrial
products, including polymers, fibbers, industrial oils and packaging materials as well as
basic chemical building blocks (green chemistry); (iii) fuels such as ethanol and biodiesel;
(iv) molecules with pharmaceutical properties and health benefits. To reach these new
agricultural perspectives, new varieties with the appropriate properties need to be selected
(Tester & Langridge, 2010) through plant breeding, be it conventional, marker assisted, QTL
mapping assisted, or genetically modified (GM) (Mittler & Blumwald, 2010). There are also
growing demands for germplasm adapted to deal with changing climates and effective
under a range of cultural practices and for foods with higher nutritional value. To decipher
agronomical traits, functional genomics approaches can be of good use to understand
physiological, molecular and genetic processes underlying complex traits. Appropriate
functional genomics technologies such as transcriptomics, proteomics and metabolomics
must be used together with detailed physiological and environmental information as a
combined platform for ‘candidate’ gene identification or translational genomics approaches
that aims to improve complex traits in plants (Sanchez et al., 2011). Without a
comprehensive understanding of the plant physiology, molecular processes and genetics of
214 Metabolomics

the components of complex traits, the development of new varieties will remain an
empirical yet uncertain procedure. This integration of functional genomics data can be
viewed as the first step to systems and predictive biology serving agricultural perspectives.
Among the ‘omics’ technologies, metabolomics is one of the more recently introduced. The
term ‘metabolome’ coined in 1998 (Oliver et al., 1998) refers to the richly diverse population
of small molecules present in biofluids, living cells or organisms. Overall, there are two
approaches to analyse small molecules, and they differ in the number of compounds
analysed, the level of structural information obtained, and their sensitivity. The most
common approach, metabolite profiling, is the analysis of small numbers of known
metabolites in specific compound classes (e.g. sugars, amino acids or phenolics). At the
other extreme, metabolic fingerprinting detects many compounds but their structures are
rarely identified. Today metabolomics methods typically allow measuring hundreds of
compounds, with a small number being definitively identified, a larger number being
identified as belonging to particular compound classes, and many remaining unidentified.
Over the past decade, metabolomics has gone from being just a simple concept to becoming
a rapidly growing discipline with valuable outputs in plant biology (Hall, 2006; Saito &
Matsuda, 2010; Hall, 2011a; Shepherd et al., 2011). Metabolomics has played a key role in
basic plant biology and started having a potentially broad field of applications. Plants
produce an astonishing wealth of metabolites estimated to figures ranging from 200,000 to
1,000,000 metabolites (Dixon & Strack, 2003; Saito & Matsuda, 2010). The first significant
advances have been made in the area of analytical technology for metabolite identification
in order to increase our capacity to simultaneously analyse a chemically diverse range of
metabolites in complex mixtures. The metabolomics community has set up analytical
platforms with complementary analytical technologies (Moing et al., 2011) after having
realized that no single technology currently available (or likely in the close future) will be
able to detect all compounds found in living cells. Today these analytical platforms provide
a combination of multiple analytical techniques such as gas chromatography (GC), liquid
chromatography (LC) or capillary electrophoresis (CE) coupled to mass spectrometry (MS),
or nuclear magnetic resonance spectroscopy (NMR) and much more (Kim et al., 2011; Lei et
al., 2011).
Considering metabolomics as a combination of knowledge and know-how in
biochemistry, signal processing, data and metadata handling, and data mining, the
challenge remains to perform in a cohesive and coordinated manner these
multidisciplinary approaches to solve biological questions (Ferry-Dumazet et al., 2011;
Hall, 2011b). Recently, plant biologists have used metabolomics approaches to understand
fundamental plant processes (Leiss et al., 2010; Sulpice et al., 2010), to make a link
between genotype and biochemical phenotype and to study plant responses to biotic or
abiotic stresses by combining genomics and biochemical phenotyping capabilities
(Redestig & Costa, 2011; Villiers et al., 2011). While full genome sequence annotations of
the major crops have been published, many post-genomic studies using metabolomics
approaches have tried to bridge the phenotype-genotype gap in order to link gene to
function (Smith & Bluhm, 2011). Such integrated approaches have been helpful in
assigning functions to a large class of function-unknown genes and their interactions with
other pathways and also useful in applications such as metabolic engineering (Liu et al.,
2009) and assessment of GM plants (Kusano et al., 2011b).
New Opportunities in Metabolomics and Biochemical Phenotyping for Plant Systems Biology 215

As part of a more recent emerging area, robust data generated from metabolomics can be
combined with computationally-intensive approaches based on modelling of pathways to
steer this field towards systems biology, which promises to provide an integrated view of
cellular processes (Joyce & Palsson, 2006; Wang et al., 2006). Bringing metabolomics data
into the forefront of system biology is a challenging opportunity that implies using
quantitative metabolomics data in the context of models to improve our understanding of
metabolism and drive the biological discovery process. So far, computational studies on
metabolomics data have often been restricted to multivariate statistical analyses such as
principal component analysis or PLS discriminant analysis to look at trends among different
data sets. Such work has proven useful in discovering potential biomarkers of stress and
identifying key metabolic difference in GM plants, but provides minimal insight into the
underlying biology or the means to modulate it for agronomic or industrial purposes. Now
researchers are rising to the challenge by using omics data integration and specially high-
throughput metabolomics data within a constraint-based framework to address
fundamental questions that would increase our understanding of systems as a whole.
This article provides an overview of the technological trends in plant metabolomics to
optimize the characterization of a large number of metabolites with accurate and absolute
quantification in a few samples (concept of vertical high-throughput metabolomics) and
present the needed technologies to increase the analysis capacity of samples for large-scale
studies (concept of horizontal high-throughput metabolomics). This article also outlines
how these technological developments in plant metabolomics can be used for systems
biology, quantitative genetics and the emerging field of meta-phenomics to answer the key
challenges of plant biology and agriculture in the future, and which technological and
computational developments are necessary to meet these challenges.

2. Technological trends in plant metabolomics


For plant metabolomics, the analytical strategies reviewed a few years ago (Weckwerth,
2007) are still widely used. Major improvements over the past five years have targeted
spectra resolution and processing (http://www.metabolomicssociety.org/software.html),
and the emergence of databases (http://www.metabolomicssociety.org/database.html).
Thanks to technological and methodological progress, numbers of analytes and compound
families that can be determined in a given sample are still increasing, but usually at the
expense of the number of samples that can be analysed due to increasing costs and/or
labour (Fig. 1). Conversely, novel experimental strategies produce increasing numbers of
samples. Thus, not only the best compromise between analyte number (vertical high-
throughput approach) and sample throughput (horizontal high-throughput approach) has
to be found, but also synergisms between such approaches.

2.1 Vertical approaches


Vertical high-throughput approaches, also called high-density approaches, are defined as
strategies that promote sample variables over sample numbers. They are especially
interesting for studies in plants given their enormous metabolic diversity. In the plant
kingdom, the species number is estimated between 270,000 (observed) and 400,000 and
the number of metabolites produced between 200,000 and 1,000,000 (Dixon & Strack, 2003;
216 Metabolomics

Saito & Matsuda, 2010). Even the number of primary metabolites, defined as the type of
compounds synthesized by all or most plant species, may exceed the number of
compounds found in other eukaryotes since plants are true autotrophs (Pichersky &
Lewinsohn, 2011). In addition, different plant lineages synthesize distinct sets of
“specialized metabolites”, often mis-named “secondary metabolites” (Pichersky &
Lewinsohn, 2011), with Arabidopsis thaliana estimated to make up to 3,500 of such
specialized metabolites. Capturing such diversity is one of the challenges for plant
metabolomics compared to animal metabolomics, which has to deal with ‘only’ 5,000 to
25,000 different metabolites (Trethewey, 2004). However, the consumption of plant-
derived food is known to lead to a strong increase in metabolite diversity in animal or
human derived samples, e.g. blood or urine. This implies that plant and nutrition
scientists face a similar challenge. Indeed, specific plant metabolites are attracting
attention due to their role/impact on health and nutrition. Vertical metabolomics mainly
relies on sophisticated instrumentation such as NMR and MS, with or without
hyphenation of chromatography or capillary electrophoresis (LC-NMR, LC-SPE-NMR,
LC-MS, GC- MS, GC- SPE-MS, CE-MS, Fourier Transform-MS (FT-MS), Table 1).

Fig. 1. Complementarities of high-throughput vertical and horizontal biochemical


phenotyping. Costs and/or labour requirements are considered similar for each technology.
New Opportunities in Metabolomics and Biochemical Phenotyping for Plant Systems Biology 217

Metabolite classes Typical metabolites Instruments


Amino acids and Amino acids, beta-alanine, GABA, CE-MS, GC-MS (after
their derivatives oxoproline derivatization), LC-MS, NMR
Amines Polyamines (putrescine, spermine, CE-MS, GC-MS, NMR
spermidine)
Betaines, choline NMR, LC-MS
Organic acids in TCA cycle intermediates CE-MS, GC-MS (after
central metabolism derivatization), NMR, LC-MS
(partially)
Other organic acids Quinic acid, shikimic acid NMR, LC-MS,
GC-MS (after derivatization), CE-
MS
Sugars and their Mono-, di- and trisaccharides, GC-MS (after derivatization), CE-
derivatives sugar alcohols, sugar mono- and MS (sugar phosphates), CE-PDA
diphosphates (partially), NMR
Phytic acid NMR, LC-MS
Alkaloids Polar alkaloids (e.g. pyrrolizidine LC-MS, NMR
alkaloids)
Fatty acids and their Saturated and unsaturated GC-MS (after derivatization)
derivatives aliphatic monocarboxylic acids
and their derivatives
Polar lipids Phospholipids, mono-, di-, and LC-MS
triacylglycerols
Isoprenoids Terpenoids and their derivatives GC-MS (non-polar), LC-MS (polar)
Nucleic acids and Purines, pyrimidines, mono-, di-, CE-MS, GC-MS (partially), NMR
their derivatives and triphosphate nucleosides
Pigments Carotenoids, chlorophylls, LC-PDA, LC-MS
anthocyanins
Volatiles Phenylpropanoid volatiles, GC-MS, GCxGC-MS
aliphatic alcohols, aldehydes,
ketones
Other specialized Polar phenylpropanoids (e.g. LC-MS, LC-(SPE)-NMR, NMR
metabolites chlorogenic acids), flavonols
Phytohormones (e.g. auxins) LC-MS

Table 1. A selection of examples of plant primary and specialized metabolites detected by a


variety of analytical techniques. Adapted from (Kusano et al., 2011a). PDA, photodiode array
detection; GABA, gamma-aminobutyrate; TCA, tricarboxylic acid cycle; SPE: solid-phase
extraction. See (Saito & Matsuda, 2010) to have an overview of plant metabolomics pipelines.
218 Metabolomics

Vertical approaches have to deal with a wide variety of chemical structures, which implies
wide ranges of solubility, polarity and stability, as well as a high dynamic range of
metabolite concentrations (>1012, (Sumner, 2010); 106, (Saito & Matsuda, 2010)). In addition,
plant metabolites are usually extracted with sometimes sophisticated protocols including
steps like heating or fractionation that may lead to losing or modifying metabolites, but also
promote the synthesis or import of chemical artefacts. This is why the term analyte, which
might be a metabolite or an artefact, is preferred. For example, during the derivatization
process, which is required for non-volatile compounds when performing GC-MS, a single
metabolite may produce multiple derivatives leading to different peaks. Similarly, adducts
and product ions are formed during the desolvation step following the ionization process in
LC-MS analyses (Werner et al., 2008). To cover the wide range of chemical diversity and
concentrations of plant metabolites, careful experimental design is definitely required,
including special care for harvest (Ernst, 1995), several extraction protocols and multi-
analytical platforms (see (Ryan & Robards, 2006; Allwood et al., 2011) and Tables 1-2).

Plant Species Analytical instruments References


Arabidopsis NMR, GC-MS, CE-MS, LC-MS, (Beale & Sussman, 2011)
DI-FT-MS review
Aspen GC-MS (Bylesjo et al., 2009)
Broccoli NMR (Ward et al., 2010)
Grape GC-MS (Deluc et al., 2007)
NMR (Mulas et al., 2011)
Tomato NMR, GC-FID, LC-MS (Mounet et al., 2009)
NMR, GC-MS, LC-MS, LC-FT-MS (de Vos et al., 2011) review
HRMAS-NMR (Sanchez-Perez et al., 2010)
GC-MS, LC-MS, CE-MS (Kusano et al., 2011b)
Maize NMR (Cossegal et al., 2008; Broyart
et al., 2010)
GC-MS (Skogerson et al., 2010)
Medicago LC-MS (Farag et al., 2008)
Melon NMR, GC-MS, LC-MS (Moing et al., 2011)
Palm trees NMR, GC-FID (Bourgis et al., 2011)
Potato GC-MS (Urbanczyk-Wochniak et al.,
2005)
Strawberry LC-MS, DI-MS (McDougall et al., 2008)
GC-MS, LC-MS (Fait et al., 2008)
Rice GC-MS, CE-MS, CE-DAD, FT-MS (Oikawa et al., 2008)
Vanilla NMR (Palama et al., 2009)
Medicinal species NMR, LC-MS, GC-MS, HPLC (Okada et al., 2010)
Table 2. Representative examples of model-, crop- and medicinal-plant metabolomics
studies using different analytical platforms. DI : Direct Infusion. FID, Flame Ionization
Detection. HRMAS-NMR: High resolution Magic-Angle Spinning NMR. ICR: Ion Cyclotron
Resonance.
New Opportunities in Metabolomics and Biochemical Phenotyping for Plant Systems Biology 219

Currently the number of quantified analytes in a given sample and in one shot is
approximately 50 with proton NMR, 100-200 with GC-MS, >1000 with LC-High-Resolution-
MS (LC-HR-MS). This expansion of scale has been made possible through improved
analytical capabilities, dissemination of routine procedures between laboratories, but also
implementation of dedicated statistical and data mining strategies. However, a large
proportion of the analytes detected in plant extracts cannot be annotated and identified
based on chemical shift and multiplicity for NMR analysis, or on elemental formula (based
on m/z ratio and isotopic ratio) and chromatographic retention time for GC- or LC-MS
analysis, alone. Hence metabolite identification, which uses a variety of analytical
techniques along with analyte/metabolite databases, remains difficult (Moco et al., 2007).
Achieving standardization for naming compounds at the plant metabolomics community
level is also an important issue, as it will enable researchers to share knowledge and speed
up metabolite identification (Saito & Matsuda, 2010; Kim et al., 2011). Another challenge is
the development of chimiotheques, where trusted reference compounds would be available
for the community to validate analyte identifications, for example via spiking experiments.

2.1.1 Optimization and combination of the different current techniques


As already mentioned (see Tables 1-2), a combination of different current techniques is
needed to cover the wide diversity of metabolites found in plants. Thus, a combination of
different MS technologies is helpful for identification purpose. Among HR-MS technologies,
LC- Time-Of-Flight (TOF) (resolution of 8,000-20,000, accuracy 1-5 ppm), Orbitrap®
(resolution of 100,000, accuracy< 1 ppm) and FT-ICR-MS (resolution> 100,000, accuracy< 1
ppm) are currently the most powerful ultra-high resolution (UHR) mass spectrometers.
They provide molecular formula information, thus offering great possibilities in terms of
metabolite identification (see (Werner et al., 2008) for the strategy, pitfalls and bottleneck of
metabolite identification). Nevertheless, the poor reproducibility and fragmentation
variability between instruments from the same brand require a home-made metabolite
database for each instrument. In addition it should be kept in mind that plant extracts
contain many isomers, i.e. with identical elemental compositions and accurate masses. UHR-
MS analysis of a selection of extracts may help to identify marker-metabolites revealed using
HR-MS on a larger range of samples. Besides, multidimensional separation techniques have
emerged in order to enhance metabolite coverage in the GC-MS (Gaquerel et al., 2009;
Allwood et al., 2011) and LC-MS (Lei et al., 2011) fields. Further methodologies such as Ion
Mobility MS (Dwivedi et al., 2008), which have not been tested in plants so far, might also
prove useful. Anyway, processing and integrating data still remain the major bottlenecks
and thus the most labour intensive steps for all these different analytical platforms.
Developments are nevertheless underway to automate them (see (Redestig et al., 2010) for
MS and hyphenated technologies).
2.1.2 From relative to absolute quantification of biological variability
Whereas the “convenient” relative quantification is often used in MS studies, absolute
quantification will be increasingly required. For example, various modelling approaches
require precise concentrations of metabolites. Furthermore, the sharing and integration of data
obtained on different analytical platforms will be greatly facilitated if expressed as absolute
quantities. To face the challenge of quantification, in GC-MS and LC-ESI-MS impaired by ion
suppression or enhancement and matrix effects, a solution is to use stable-isotopomers of
220 Metabolomics

target metabolites or to run whole 13C metabolome isotope labelling (Feldberg et al., 2009;
Giavalisco et al., 2009). However, even with a stable isotope the matrix effects may impair the
quantification (Jemal et al., 2003) and few isotopically-labelled metabolites are currently
commercially available (Lei et al., 2011). In contrast to MS-based technologies, NMR, although
less sensitive, provides ease of quantitation since the resonance intensity is only determined by
the molar concentration, and high reproducibility (Ward et al., 2010; Kim et al., 2011).
Surprinsingly, a unique extraction protocol (sometimes one-step protocol) is typically used for a
given analytical technique, regardless of the vast variety of plant matrices (plant species, organs
and tissues). Very few metabolomic publications are prolix on extraction recovery and stability.
Running blanks (solvent blank and extraction blank) in the same conditions as the biological
samples is also important, as it is needed to identify impurities originating from solvents (Kaiser
et al., 2009) or consumables (i.e., phthalates from plastic ware) (Allwood et al., 2011; Weckwerth,
2011). Although metabolomics is by definition an untargeted approach, highly selective
extraction protocols along with targeted analysis should not be forgotten, especially to reach
high and reproducible extraction recovery as well as quantification accuracy (Sawada et al.,
2009). Then, replication is required to achieve statistical reliability. Biological replicates should
be preferred to technological replicates assuming biological variance almost always exceeds
analytical variance (Shintu et al., 2009). Five biological replicates of five pooled-tissue samples
or of five individuals and two to three technological replicates are recommended in plant
metabolomics to get statistically reliable information (Tikunov et al., 2007). Quality control
samples should also be run (Fiehn et al., 2008; Allwood et al., 2011).

2.2 Horizontal approaches


Horizontal approaches are defined as strategies that promote sample number over number
of variables being measured. Mutant screens and quantitative genetics are typical examples
requiring horizontal high-throughput, as they typically involve experiments with hundreds
to thousands of samples. Targeted assays are usually preferred due to their low needs in
terms of labour and/or costs, although several untargeted strategies such as bucketing and
fingerprinting are also amenable to very high numbers of samples. While the processing of
raw data still represents the slowest step in vertical strategies, the toughest bottleneck in
horizontal high-throughput approaches is probably sample logistics.

2.2.1 Sample logistics


In large scale experiments, harvesting, grinding and weighing become extremely work
intensive (>75% of the time), especially when samples need to be kept at very low
temperatures to avoid alteration of their biochemical composition. Due to the highly
dynamic nature of the metabolome, harvesting and quenching of samples into liquid
nitrogen should also be achieved as quickly as possible (Ap Rees et al., 1977). Unfortunately,
fast solutions are very limited (e.g. leaf punchers), and thus recruiting as many as possible
helpers probably remains the best way to achieve a reliable large-scale harvest. Sample
storage may also become problematic when sample turnover dramatically increases. A good
way to avoid losses of samples and overfilling of -80°C freezers is to use software enabling
sample management. Although costly, available automated storage solutions may also
dramatically improve the handling of samples.
New Opportunities in Metabolomics and Biochemical Phenotyping for Plant Systems Biology 221

Most analytical technologies require sample grinding prior to extraction and analysis. Mills
enabling the parallel grinding of large numbers of samples (e.g., 192 samples) are now
available at affordable prices. However, they usually do not allow multiparallel grinding of
samples of large size, suggesting that further developments are needed to enable large-scale
studies with organs such as fruits or ears and with most crops. Last but not least, the
weighing of aliquots is a tedious task, especially when the material needs to be kept at very
low temperature. A robot combining grinding and weighing of up to 96 samples has been
developed recently (http://www.labman.co.uk), opening the way for unprecedented
horizontal high-throughput.

2.2.2 Microplate technology


The first microplate was fabricated in 1951 by the Hungarian Gyula Takátsky (Takatsy,
1955). It was made of 72 wells machined in a polymethyl methacrylate block and was used
to speed up serial dilutions. This invention was driven by the need for a fast and reliable
diagnostic for influenza, as Hungary was facing a major epidemic at that time. Sixty years
later, the microplate format has driven the development of a huge diversity of labware and
equipment, and hundreds of millions of microplates are sold every year. Sample storage,
extractions and dilutions can be achieved in microplates, which remain the fastest and
cheapest solution to process large numbers of samples in parallel. The quantification of
various metabolites can be achieved in microplates via chemical or enzymatic reactions
yielding products that can be quantified in a wide range of dedicated readers. The most
common and cheapest readers are filter-based UV-visible spectrophotometers. They enable
the quantification of a wide range of metabolites, including major sugars, organic acids and
amino acids using endpoint methods (Bergmeyer, 1983, 1985, 1987), and metabolic
intermediates that are present at much lower concentrations using kinetic assays (Gibon et
al., 2002). Fluorimetry (Hausler et al., 2000) and luminometry (Roda et al., 2004) also provide
high sensitivity and benefit from many commercially available fluorigenic substrates. Their
use is nevertheless restricted in plants, due to the quenching of the emitted light that occurs
in the presence of e.g. polyphenols that are usually present in plant extracts.
Throughput on microplates can be dramatically increased by using pipetting robots, which
can handle up to 1536 samples in parallel and down to the nanoliter scale, depending on the
brand. Thus, using one 96-head robot equipped with microplate handling and a series of
microplate readers, a single person can run the determination of a given metabolite in
thousands of samples per week. Increasing the number of analytes would nevertheless
result in a decrease in sample throughput, roughly by a factor 2 at each supplemental
analyte. It is estimated that at equal costs, such an approach might be of advantage for 10 to
20 analytes over other targeted technologies such as LC-MS/MS, which have already
proven efficient for the capture of relatively large numbers of metabolites from the same
class at high-throughput (Rashed et al., 1997) and 2.2.3 Section below. Microplates of
increasing density formats (up to 9600 wells per plate) have been released to increase the
overall throughput of analyses and decrease the costs per assay. Such miniaturisation
nevertheless faces physical constraints of delivering very small volumes to wells and of
detecting responses in a manner that is both sensitive and rapid (Battersby & Trau, 2002).
The use of volumes in the nanoliter range is also limited by quick evaporation of the solvent
used for analysis. A further drawback is high costs in terms of equipment (e.g., pipetting
222 Metabolomics

robots and readers able to handle high density plates), which implies that very high
numbers of samples will have to be processed before decreasing costs per assay. These
limitations probably explain why the use of high density microplates has not been adopted
by a wide research community so far.

2.2.3 Targeted MS technologies: Quantification of selected biochemical markers


using LC-MS
Targeted analysis for small molecules using MS may use different technologies: GC-MS
(Koek et al., 2011) , CE-MS (Ramautar et al., 2011), LC-MS and more recently MALDI-MS
(Shroff et al., 2009). Here only LC-MS will be dealt with. LC-MS technology has been used
for quantification long before the ages of the "omics". Despite its high-skilled technical need
and its expensive cost, it has gained popularity in the metabolomics field. Triple
quadrupoles analyzers (TQMS) are the workhorse of LC-MS quantification. They are mostly
operated in multiple reaction monitoring (MRM) mode to achieve high selectivity and
sensitivity. A new promising approach is the use of high resolution extracted ion
chromatograms from full scans of high resolution instruments (Lu et al., 2008). Main
advantages over MRM are the virtually unlimited number of monitored compounds and the
possibility to reanalyze data after acquisition by extracting ion chromatograms
corresponding to new compounds of interest.
Calibration of these methods involves most of the time internal calibration, with or without
use of stable isotope analogs (Ciccimaro & Blair, 2010). For instance, quantification of amino
acids by LC-MS (MRM) in barley was calibrated using d2-Phe as an internal standard. The
interday precision of the method ranged from 3.7 to 9.4 % RSD, depending on the amino-
acid (Thiele et al., 2008). However an isotopic dilution calibration is not always possible due
to the lack of the corresponding labelled metabolite or its cost. These targeted LC-MS
methods must undergo a complete method validation. They need fast separation, high
selectivity, linearity range and limits of quantification in agreement with the metabolite
level. For methods involving atmospheric pressure ionization, a careful evaluation of matrix
effect on quantification and its minimization should be addressed (Trufelli et al., 2011).
Moreover, to be relevant, these methods must obviously be applied after an exhaustive
extraction evaluated by recovery procedures.
Targeted approaches have been applied in functional genomics. A "widely targeted"
metabolomics approach based on LC-MS (MRM) has been proposed (Sawada et al., 2009). It
consisted in repeated UPLC-TQMS analyses performed on a same sample. Each 3 min
analytical run allowed simultaneous detection of 5 compounds. Expected throughput was
estimated 1,000 biological samples per week for quantification of about 500 metabolites. This
methodology was later applied on mature seeds of 2656 mutants and 225 Arabidopsis
accessions for 17 amino-acids, 18 glucosinolate derivatives and one flavonoid, leading to
characterization of amino-acids hyper-accumulating genotypes (Hirai et al., 2010). They
have also been applied in phytochemistry and phytomedicine. For instance, Chinese
medicinal herbs were tested for two secondary metabolites inducing nephrotoxicity. The
UPLC-MS (MRM) run was 5 min and was amenable for high-throughput analyses (Jacob et
al., 2007) This method was however impaired by a strong matrix effect that could not be
prevented and another approach was preferred. At last, these techniques have been also
shown to be a must for some classes of compounds such as hormones (Kojima et al., 2009),
New Opportunities in Metabolomics and Biochemical Phenotyping for Plant Systems Biology 223

intermediates of central metabolism (Arrivault et al., 2009) and pesticides (Kmellar et al.,
2010). In fact, they provide together the appropriate selectivity, sensitivity and throughput.

2.2.4 Further technologies


Other technologies involve miniaturization of the separation step used prior to detection. A
key step in miniaturization and automation of chromatography is the development of
microfluidic systems, which process or manipulate very small volumes (down to 10-18 L)
using channels of micrometre dimensions (Whitesides, 2006). The fact that factors such as
surface tension and viscosity are getting very different in such systems brings many new
possibilities to control concentrations and behaviours of molecules, particles or even cells
(Nagrath et al., 2007) in space and time. Thus, the performance of soft lithography on e.g.
poly(dimethylsiloxane) (McDonald et al., 2000) or polypropylene (Vengasandra et al., 2010)
enables the design of reservoirs, channels, valves, and reaction chambers that can be used to
separate and transform a wide range of molecules. Combined to detection systems such as
laser induced fluorescence (Jiang et al., 2000), infrared spectroscopy (Shaw et al., 2009) or
electrochemical electrodes (Eklund et al., 2006), they are well suited for massively parallel
assays and provide the advantage of using very small amounts of reagents and samples.
Further advantages are high resolution and sensitivity as well as fast analysis.
The use of microfluidic systems for metabolite analysis has just begun. Whereas applications
targeting one molecule, e.g. glucose (Atalay et al., 2009), have been developed, the possibility
to separate molecules has already enabled the profiling of classes of metabolites such as
glucosinolates (Fouad et al., 2008) or flavonoids (Hompesch et al., 2005). Furthermore, the ease
of creating systems able to distribute fluids into multiple channels enables the performance of
several assays in parallel (Moser et al., 2002), or even n-multidimensional separations (Tomas
et al., 2008) that would eventually be coupled to various detection devices, opening
unprecedented possibilities for targeted and untargeted metabolomics.
Unfortunately, microfluidics have not yet benefited from standardisation, which hampers
their adoption by a wide research community. Besides involving complex designs and
fabrication techniques prohibiting widespread use due to cost and/or time for production,
microfluidic systems may require unfamiliar laboratory habits. Therefore, one logical next
step is the integration with the standardized microplate layout, thus taking advantage of the
extensive work of the lab automation community (Choi & Cunningham, 2007; Halpin &
Spence, 2010). Strikingly, such integration might ultimately result in methodologies
enabling high density analyses on very large numbers of samples, thus breaking the
relationship depicted in Figure 1. Finally, microfluidics and more generally
nanotechnologies have almost certainly much more to offer, as future developments could
for example lead to portable systems that would allow metabolite profiling directly in the
field, thus shortcutting sample handling, or even to chips embarked on growing plants that
would be able to monitor fluxes in situ.

2.3 Complementarities of vertical and horizontal approaches


The combination of horizontal and vertical high-throughput approaches (Fig. 1) is of
particular interest, as it has the potential to dramatically speed up the process of discovery.
Thus, depending on the objectives of the study, an untargeted approach can be used first on
224 Metabolomics

a selection of samples to identify the most discriminating biomarkers that would then be
analyzed on a much greater number of samples using a targeted approach (Tarpley et al.,
2005). For example, such strategy has been successfully used in maize where a number of
enzymes were first profiled in a small panel of eight highly diverse maize inbred lines,
revealing a highly heritable variation in NAD-dependent isocitrate dehydrogenase activity.
The use of a panel of about hundred lines then allowed the identification of a novel amino-
acid substitution in a phylogenetically conserved site, which is assoaciated with isocitrate
activity variation (Zhang et al., 2010). On the contrary, a horizontal approach can be used to
screen large numbers of samples, thus revealing the most extremes or representative ones,
on which a vertical approach can then be used to search for unexpected modifications, to
study the system as a whole in the best possible matrix of samples, or simply to find novel
biomarkers. As an example, the easy to measure glucose-6-phosphate, which is a good
temporal marker of carbon depletion (Stitt et al., 2007), has been used to define a precise
time frame to study transcriptomic and metabolomic responses to carbon starvation in
Arabidopsis leaves (Usadel et al., 2008), thus avoiding unnecessary and costly analyses.

3. Key challenges for plant metabolomics


An increasing number of approaches benefit from plant metabolomics. Among them,
systems biology, quantitative genetics and meta-phenomics offer particularly exciting yet
challenging perspectives.

3.1 Systems biology


In the context of plant functional genomics, the combination of metabolomics, proteomics,
and transcriptomics has permitted to decipher and understand dynamic interactions in
metabolic networks and to discover new correlations with biochemically characterized
pathways as well as pathways hitherto unknown (Zhang et al., 2009; Williams et al., 2010).
The main lesson from the latter or similar studies is that metabolic pathways are highly
interactive rather than operating as separate units. In each biological system (cell, tissue,
organism) there are metabolic networks in place, which are highly flexible and present a
huge capacity to provide compensatory mechanisms through regulatory process. These
observations actually explain why many dedicated GMO strategies ended up with silent
phenotype (Weckwerth et al., 2004) and also convinced a large set of researchers to study
metabolic networks as a whole, and not as a sum of parts, moving from reductionist to
holistic approaches.
Although systems biology may mean different things to different people, there is a common
understanding that this discipline is a comprehensive quantitative analysis of the manner in
which all the components of a biological system (cell, tissue, organisms, communities)
interact functionally over time. Systems biology aims at combining omics data resulting
from complex networks into computational models. Besides integration with upstream
levels (genome, transcriptome, proteome), metabolite data also have to be integrated with
downstream levels (e.g. growth, performance) data. The quantitative data are the initial
point for the formulation of mathematical models, which are refined by hypothesis-driven,
iterative systems perturbations and data integrations. Cycles of iteration result in a more
accurate model and ultimately the model explains emergent properties of the biological
New Opportunities in Metabolomics and Biochemical Phenotyping for Plant Systems Biology 225

system of interest. Once the model is sufficiently accurate and detailed, it allows biologists
to accomplish two tasks (1) predict the behavior of the system given any perturbation such
as a modification of the environment, and (2) redesign or perturb the gene regulatory
network to create completely new emergent systems properties (Vidal, 2009; Westerhoff et
al., 2009; Arkin & Schaffer, 2011).
Exciting examples of integrated system biology to solve biological questions in plant science
have been published such as identification of key players in the branched amino acid
metabolism in A. thaliana (Curien et al., 2009), analysis of carbohydrate dynamics during
acclimation to low temperature in A. thaliana (Nagele et al., 2011), or understanding the
metabolism of tobacco grown on media containing different cytokins (Lexa et al., 2003).
Systems biology will benefit from close collaborations between different teams covering
complementary sectors of metabolism, e.g. central metabolism and different sectors of
secondary metabolism. The challenges in establishing such systems approaches rely on
collecting reliable, quantitative and systemic “omics” data, including metabolomics data, for
developing modelling able to predict de novo biological outcomes given the list of the
components involved. Advances in plant genome sequencing, transcriptomics and
proteomics have paved the way for a systematic analysis of cellular processes at gene and
protein levels. For metabolomics, some limitations remain for real system biology
approaches, in terms of analytical sensitivity, throughput and access to specific tissue or
subcellular compartments. Moreover, the high turn-over rate of many metabolic
intermediates has to be taken into consideration. In addition, the absolute quantification of
metabolites under physiological, in vivo and dynamic conditions remains a major challenge.
The combination of existing multiparallel analytical platforms with special attention to
metabolite quantification (see Sections 2.1.2 and 2.2.3) in a cohesive manner may not be
sufficient and emerging microtechnologies such as microfluidics will certainly help (see
Section 2.2.4 and (Wurm et al., 2010)).
Recently, plant systems biology has been redefined from cell to ecosystem (Keurentjes et al.,
2011). For these authors, in a holistic systems-biology approach, plants have to be studied at
six levels of biological organization (from subcellular level to ecosystem) in an orchestrated
way, with special attention to the interdependence between the various levels of biological
organization. The corresponding challenge will be to generate accurate experimental data
for communities, populations, single whole plants, down to cell types and their organelles
that can be used to feed new modelling concepts. For example, at the subcellular level
molecular signaling pathways are crucial to understand cell development, defense against
pathogens and many more intermediate processes in plants. The highly sensitive and high-
throughput method developed for the simultaneous analysis of 43 molecular species of
cytokinins, auxins, ABA and gibberellins (Kojima et al., 2009) has opened a big opportunity
to routinely describe basic molecular signaling pathways in plant cells. Others challenges
need to be considered in terms of dry labs. Because systems biology heavily relies on
information stored in public databases for the different levels of biological organization,
which is often incomplete, not standardized or improperly annotated, it is essential that
collective efforts are developed for the validation of large data sets. Plant network biology is
in its infancy and other current needs range from the development of new theoretical
methods to characterize network topology, to insights into dynamics of motif clusters and
biological function.
226 Metabolomics

3.2 Quantitative genetics


Quantitative genetics, which aim at associating quantitative traits with genomic regions
called quantitative trait loci (QTL), represent a great opportunity to understand the diversity
of plant metabolism and its relationship to nutritional value or biomass production. Studies
combining metabolomics and quantitative genetics performed in Arabidopsis seedlings
(Keurentjes et al., 2006) and tomato fruits (Schauer et al., 2006) have shown that variations in
metabolite levels are for a large part heritable, and have identified large numbers of
metabolite QTL, implying that levels of metabolites of interest could be controlled by
manipulating small genome regions (Saito & Matsuda, 2010). Conversely, genetic diversity
has been used to study the behaviour of metabolic networks and the way they integrate
with whole plant traits, eventually revealing links between metabolic composition and
growth (Meyer et al., 2007). Such findings appeal for multivariate QTL mapping (Calinski et
al., 2000), thus opening exciting perspectives for the manipulation of plant performance.
The identification of the molecular bases underlying QTL has usually been a major
challenge, and several years of hard work were typically necessary to unravel just one of
them. However, thanks to the development of increasingly powerful methodologies
exploiting genetic diversity that combine linkage and/or association mapping and high
density genotyping, the elucidation of such molecular bases can now be achieved much
quicker (Myles et al., 2009). The other side of the coin is that these methodologies require
experiments of increasing sizes. Thus, the nested association mapping (NAM) approach
recently developed in maize (Yu et al., 2008) already involves 5,000 genotypes (25 mapping
populations of 200 genotypes each), which would represent at least 5,000 samples to
process. Unfortunately, due to technical and financial limitations, the processing of so many
samples remains very unusual in plant metabolomics. Furthermore, taking into account
different growth scenarios, temporal aspects or different organs or tissues would result in
factorial increases in numbers of samples. As mentioned above, combinations of horizontal
and vertical metabolomics might nevertheless be very useful to decrease costs and labour.
For instance, small sub-panels with high genetic diversity can be used first to assess
heritability for a large number of metabolic traits, selected ones being then evaluated in full
panels using inexpensive and fast methods.
Finally and importantly, genetic divergence and phenotypic divergence are too different
things (Kozak et al., 2011). Accordingly, one single gene can be responsible for huge
phenotypic variations and one single trait can be controlled by many QTL. One consequence
is that molecular marker-assisted breeding might not always be the best and/or cheapest
solution to select genotypes yielding phenotypes of interest. Therefore, it is pertinent to
explore the possibility to use alternative biomarkers, including metabolites that can be
measured at reasonable costs in very large populations.

3.3 Meta-phenomics
Comparing different species is a powerful way to extend knowledge about biological
processes. Thus, comparative genomics facilitate the assignation of gene function in non
sequenced organisms, enable the quick annotation of newly sequenced genomes and greatly
contribute to studies of gene function and evolution. For example, extensive synteny
between genomes of Graminae species has been shown (Salse, 2004) and QTL controlling
New Opportunities in Metabolomics and Biochemical Phenotyping for Plant Systems Biology 227

similar traits have been found in orthologous regions of e.g., maize and sorghum
(Figueiredo et al., 2010). Conversely, the fact that orthologous genes do not necessary have
the same functions in different species (Buckler et al., 2009) opens fascinating perspectives
regarding evolution of gene function (Wang et al., 2009).
Finding common and divergent phenotypes among large numbers of species is also a
promising way to better understand biological functions in the context of evolution.
Meta-phenomics, which has recently been proposed by Poorter and colleagues (Poorter et
al., 2009; Poorter et al., 2010), defines as the study of plant responses to environmental
factors by performing meta-analyses. This novel ecophysiological approach aims at
generalising plant responses by integrating phenotypic and environmental data gathered for
large numbers of species. Thus, by using accurate normalisation procedures generic
response curves were found for surface leaf area as related to major abiotic factors.
Noteworthy, data for >300 species had to be collected and curated manually throughout 60
years of literature. One exciting finding is divergences between groups of species could be
pinpointed, for example C3 and C4 species. There is no doubt that meta-phenomics is
amenable to the cellular level, and in particular to metabolic pathways, and C3 and C4
metabotypes are indeed easy to distinguish when comparing their respective metabolomes.
However, this might be considerably complicated given the heterogeneity of available
metabolic data (in terms of e.g., annotation and normalisation). Furthermore, descriptions of
environmental conditions found in literature are almost always text-based, and thus very
difficult to compute. Fortunately, the adoption and use of standardised conceptualisations
with explicit specifications to report data and metadata (i.e. minimum checklists) is
progressing in the field of metabolomics (Fiehn et al., 2007a; Fiehn et al., 2007b). It will
nevertheless be of central importance to prefer absolute quantification and to enable
quantitative descriptions of environmental factors, which will probably be facilitated via
collaborations with ecophysiologists.

4. Conclusion
As metabolomics in general (Hall et al., 2011), plant metabolomics is moving towards
biology with a growing variety of applications from ‘simple’ diagnostic of culture practices
to translational studies towards systems biology. However, for some of the emerging
applications, the optimization of analytical and computational technologies for the
acquisition, handling and mining of metabolomics data remains necessary. Some of the
crucial bottlenecks that still have to be adressed concern quantification for modelling, time
and spatial resolved experiments, multi-experiments and data sharing.
The promotion of multi-experiments and multi-labs combined analyses (Allwood et al.,
2009; Ward et al., 2010) for high sample numbers, indispensable for some ecology or
quantitative genetics studies for instance, requires shared plant biological standards (labeled
or non-labeled) and standardization of their use. The absolute quantification data, needed
for metabolism modelling in systems biololy approaches, also requires isotopically labelled
plant standards or at least labelled reference compounds for MS approaches. The
generalisation of time-resolved experiments for instance for the study of fine metabolism
regulation or short-term responses to stresses will need further increases in horizontal high-
throughput using microplate, microfluidics or other technologies. Besides increased
throughput, increased sensitivity for all the analytical technologies listed in this review may
228 Metabolomics

open new insights into the use of metabolomics for plant development studies. Spatial-
resolved experiments with analysis of laser-microdissected samples by NMR or MS (Moco
et al., 2009; Kim et al., 2011) will be particularly useful for the study of plant-pathogen
interactions. The generalization of metabolite compartmentation studies in plant tissues at
the cellular and subcellular levels, possibly with non-aqueous fractionation (Krueger et al.,
2011), will also request increases in both horizontal high-throughput and sensitivity.
Moreover, the systematic sharing, combining, and re-exploring of the data produced using
targeted metabolic phenotyping or untargeted metabolomics will produce new knowledge.
Cataloging the metabolome itself by experimental data and literature data, stored in curated
databases can complement genomic reconstructions of metabolism (Fiehn et al., 2011).
Access to the regulation of the plasticity and flexibility of metabolic networks implies that
the metadata of each experiment, including environment metadata (Hannemann et al., 2009)
have to be carefully documented and uploaded into a central or distributed network
repository dedicated to plants. This suggests that the MSI initiative (Fiehn et al., 2007a) has
to continue to propose and promote standardization criteria that will be integrated by the
bioinformatics developments of open repositories and used by the community. In addition,
sophisticated but easy-to-use tools for metabolomics data combining, integration with other
phenotyping or omics data, and integrated statistical analyses and modelling are needed.
The plant metabolome community may benefit from more interaction with the human
metabolome community for the use and development of such tools, and both may address
combined analyses of food quality determinants (Hall et al., 2008) and food human
consumption monitoring (Wishart, 2008).

5. Acknowledgment
Financial supports of ERA-NET ERASysBio+ (FRIM) and FP7 KBBE (DROPS, grant
agreement number FP7-244374) are acknowledged. All authors acknowledge support from
the Metabolome Facility of Bordeaux Functional Genomics Centre.

6. References
Allwood, J.W.; de Vos, C.H.R.; Moing, A.; Deborde, C.; Erban, A.; Kopka, J.; Goodacre, R. &
Hall, R. (2011). Plant metabolomics and its potential for systems biology research:
background concepts, technology and methodology, In: Methods In Systems Biology,
Westerhoff, H., Hayes, N., (Eds) in press, Elsevier Inc., isbn 978-0-12-385118-5,
Amsterdam, Netherlands
Allwood, J.W.; Erban, A.; de Koning, S.; Dunn, W.B.; Luedemann, A.; Lommen, A.; Kay, L.;
Loscher, R.; Kopka, J. & Goodacre, R. (2009). Inter-laboratory reproducibility of fast
gas chromatography-electron impact-time of flight mass spectrometry (GC-EI-
TOF/MS) based plant metabolomics. Metabolomics, Vol.5, No. 4, (Dec 2009), pp.
479-496, issn 1573-3882
Ap Rees, T.A.; Fuller, W.A. & Wright, B.W. (1977). Measurements of glycolytic intermediates
during onset of thermogenesis in spadix of Arum maculatum. Biochimica Biophysica
Acta, Vol.461, No. 2, (Aug 1977), pp. 274-282, issn 0006-3002
Arkin, A.P. & Schaffer, D.V. (2011). Network News: Innovations in 21st Century Systems
Biology. Cell, Vol.144, No. 6, (Mar 2011), pp. 844-849, issn 0092-8674
New Opportunities in Metabolomics and Biochemical Phenotyping for Plant Systems Biology 229

Arrivault, S.; Guenther, M.; Ivakov, A.; Feil, R.; Vosloh, D.; van Dongen, J.T.; Sulpice, R. &
Stitt, M. (2009). Use of reverse-phase liquid chromatography, linked to tandem
mass spectrometry, to profile the Calvin cycle and other metabolic intermediates in
Arabidopsis rosettes at different carbon dioxide concentrations. Plant Journal,
Vol.59, No. 5, (Sep 2009), pp. 824-839, issn 0960-7412
Atalay, Y.T.; Witters, D.; Vermeir, S.; Vergauwe, N.; Verboven, P.; Nicolai, B. & Lammertyn,
J. (2009). Design and optimization of a double-enzyme glucose assay in
microfluidic lab-on-a-chip. Biomicrofluidics, Vol.3, No.4, (Oct-Dec 2009), issn 1932-
1058
Battersby, B.J. & Trau, M. (2002). Novel miniaturized systems in high-throughput screening.
Trends in Biotechnology, Vol.20, No. 4, (Apr 2002), pp. 167-173, issn 0167-7799
Beale, M.H. & Sussman, M.R. (2011). Metabolomics of Arabidopsis Thaliana, In: Biology of
Plant Metabolomics, Hall, R.D., (Ed.) 157-180, Wiley-Blackwell, isbn 978-1-4051-9954-
4, Chichester, UK
Bergmeyer, H.U. (1983). Metabolites 1: Carbohydrates, In: Methods of enzymatic analysis,
Bergmeyer, J., GraBl, M., (Eds) 701, VCH Verlagsgesellschaft mbH, isbn 3-527-
26046-3, Weinheim, Germany
Bergmeyer, H.U. (1985). Metabolites 2: Tri and dicarboxylic acids, purines, pyrimidines and
derivatives, coenzymes, inorganic compounds, In: Methods of enzymatic analysis,
Bergmeyer, J., GraBl, M., (Eds) 656, VCH Verlagsgesellschaft mbH, isbn 3-527-
26047-1, Weinheim, Germany
Bergmeyer, H.U. (1987). Metabolites 3 - Lipids, Amino Acids and Related Compounds In:
Methods of enzymatic analysis, Bergmeyer, J., GraBl, M., (Eds) VCH
Verlagsgesellschaft mbH, isbn 3-527-26048-X, Weinheim, Germany
Bourgis, F.; Kilaru, A.; Cao, X.; Ngando-Ebongue, G.F.; Drira, N.; Ohlrogge, J.B. & Arondel,
V. (2011). Comparative transcriptome and metabolite analysis of oil palm and date
palm mesocarp that differ dramatically in carbon partitioning. Proceedings of the
National Academy of Sciences of the United States of America, Vol.108, No. 30, (Jul
2011), pp. 12527-12532, issn 0027-8424
Broyart, C.; Fontaine, J.X.; Molinie, R.; Cailleu, D.; Terce-Laforgue, T.; Dubois, F.; Hirel, B. &
Mesnard, F. (2010). Metabolic profiling of maize mutants deficient for two
glutamine synthetase isoenzymes using (1)H-NMR-based metabolomics.
Phytochemical Analysis, Vol.21, No. 1, (Jan-Feb 2010), pp. 102-109, issn 0958-0344
Buckler, E.S.; Holland, J.B.; Bradbury, P.J.; Acharya, C.B.; Brown, P.J.; Browne, C.; Ersoz, E.;
Flint-Garcia, S.; Garcia, A.; Glaubitz, J.C.; Goodman, M.M.; Harjes, C.; Guill, K.;
Kroon, D.E.; Larsson, S.; Lepak, N.K.; Li, H.H.; Mitchell, S.E.; Pressoir, G.; Peiffer,
J.A.; Rosas, M.O.; Rocheford, T.R.; Romay, M.C.; Romero, S.; Salvo, S.; Villeda, H.S.;
da Silva, H.S.; Sun, Q.; Tian, F.; Upadyayula, N.; Ware, D.; Yates, H.; Yu, J.M.;
Zhang, Z.W.; Kresovich, S. & McMullen, M.D. (2009). The genetic architecture of
maize flowering time. Science, Vol.325, No. 5941, (Aug 2009), pp. 714-718, issn 0036-
8075
Bylesjo, M.; Nilsson, R.; Srivastava, V.; Gronlund, A.; Johansson, A.I.; Jansson, S.; Karlsson,
J.; Moritz, T.; Wingsle, G. & Trygg, J. (2009). Integrated analysis of transcript,
230 Metabolomics

protein and metabolite data to study lignin biosynthesis in hybrid aspen. Journal of
Proteome Research, Vol.8, No. 1, (Jan 2009), pp. 199-210, issn 1535-3893
Calinski, T.; Kaczmarek, Z.; Krajewski, P.; Frova, C. & Sari-Gorla, M. (2000). A multivariate
approach to the problem of QTL localization. Heredity, Vol.84, No. 3, (Mar 2000),
pp. 303-310, issn 0018-067X
Choi, C.J. & Cunningham, B.T. (2007). A 96-well microplate incorporating a replica molded
microfluidic network integrated with photonic crystal biosensors for high
throughput kinetic biomolecular interaction analysis. Lab on a Chip, Vol.7, No. 5,
(Mar 2007), pp. 550-556, issn 1473-0197
Ciccimaro, E. & Blair, I.A. (2010). Stable-isotope dilution LC-MS for quantitative biomarker
analysis. Bioanalysis, Vol.2, No. 2, (Feb 2010), pp. 311-341, issn 1757-6180
Cossegal, M.; Chambrier, P.; Mbelo, S.; Balzergue, S.; Martin-Magniette, M.L.; Moing, A.;
Deborde, C.; Guyon, V.; Perez, P. & Rogowsky, P. (2008). Transcriptional and
metabolic adjustments in ADP-glucose pyrophosphorylase-deficient bt2 maize
kernels. Plant Physiology, Vol.146, No. 4, (Apr 2008), pp. 1553-1570, issn 0032-0889
Curien, G.; Bastlen, O.; Robert-Genthon, M.; Cornish-Bowden, A.; Cardenas, M.L. & Dumas,
R. (2009). Understanding the regulation of aspartate metabolism using a model
based on measured kinetic parameters. Molecular Systems Biology, Vol.5, (May
2009), issn 1744-4292
de Vos, R.C.H.; Hall, R. & Moing, A. (2011). Metabolomics of a model fruit: tomato In:
Biology of Plant Metabolomics, Hall, R., (Ed.) 109-155, Wiley-Blackwell Ltd, isbn 978-
1-4051-9954-4, Oxford
Deluc, L.G.; Grimplet, J.; Wheatley, M.D.; Tillett, R.L.; Quilici, D.R.; Osborne, C.; Schooley,
D.A.; Schlauch, K.A.; Cushman, J.C. & Cramer, G.R. (2007). Transcriptomic and
metabolite analyses of Cabernet Sauvignon grape berry development. BMC
Genomics, Vol.8 (Nov 2007), issn 1471-2164
Dixon, R.A. & Strack, D. (2003). Phytochemistry meets genome analysis, and beyond.
Phytochemistry, Vol.62, No. 6, (Mar 2003), pp. 815-816, issn 0031-9422
Dwivedi, P.; Wu, P.; Klopsch, S.J.; Puzon, G.J.; Xun, L. & Hill, H.H. (2008). Metabolic
profiling by ion mobility mass spectrometry (IMMS). Metabolomics, Vol.4, No. 1,
(Mar 2008), pp. 63-80, issn 1573-3882
Eklund, S.E.; Snider, R.M.; Wikswo, J.; Baudenbacher, F.; Prokop, A. & Cliffel, D.E. (2006).
Multianalyte microphysiometry as a tool in metabolomics and systems biology.
Journal of Electroanalytical Chemistry, Vol.587, No. 2, (Feb 2006), pp. 333-339, issn
0022-0728
Ernst, W.H.O. (1995). Sampling of plant material for chemical analysis. Science of the Total
Environment, Vol.176, No. 1-3, (Dec 1995), pp. 15-24, issn 0048-9697
Fait, A.; Hanhineva, K.; Beleggia, R.; Dai, N.; Rogachev, I.; Nikiforova, V.J.; Fernie, A.R. &
Aharoni, A. (2008). Reconfiguration of the achene and receptacle metabolic
networks during strawberry fruit development. Plant Physiology, Vol.148, No. 2,
(Oct 2008), pp. 730-750, issn 0032-0889
Farag, M.A.; Huhman, D.V.; Dixon, R.A. & Sumner, L.W. (2008). Metabolomics reveals
novel pathways and differential mechanistic and elicitor-specific responses in
New Opportunities in Metabolomics and Biochemical Phenotyping for Plant Systems Biology 231

phenylpropanoid and isoflavonoid biosynthesis in Medicago truncatula cell cultures.


Plant Physiology, Vol.146, No. 2, (Feb 2008), pp. 387-402, issn 0032-0889
Feldberg, L.; Venger, I.; Malitsky, S.; Rogachev, I. & Aharoni, A. (2009). Dual labeling of
metabolites for metabolome analysis (DLEMMA): A new approach for the
identification and relative quantification of metabolites by means of dual isotope
labeling and liquid chromatography-mass spectrometry. Analytical Chemistry,
Vol.81, No. 22, (Nov 2009), pp. 9257-9266, issn 0003-2700
Ferry-Dumazet, H.; Gil, L.; Deborde, C.; Moing, A.; Bernillon, S.; Rolin, D.; Nikolski, M.; de
Daruvar, A. & Jacob, D. (2011). MeRy-B: a web knowledgebase for the storage,
visualization, analysis and annotation of plant NMR metabolomic profiles. BMC
Plant Biology, Vol.11 (Jun 2011), pp. 12, issn 1471-2229
Fiehn, O.; Barupal, D.K. & Kind, T. (2011). Extending biochemical databases by metabolomic
surveys. Journal of Biological Chemistry, Vol.286, No. 27, (Jul 2011), pp. 23637-23643,
issn 0021-9258
Fiehn, O.; Robertson, D.; Griffin, J.; van der Werf, M.; Nikolau, B.; Morrison, N.; Sumner,
L.W.; Goodacre, R.; Hardy, N.W.; Taylor, C.; Fostel, J.; Kristal, B.; Kaddurah-Daouk,
R.; Mendes, P.; van Ommen, B.; Lindon, J.C. & Sansone, S.A. (2007a). The
metabolomics standards initiative (MSI). Metabolomics, Vol.3, No. 3, (Sep 2007), pp.
175-178, issn 1573-3882
Fiehn, O.; Sumner, L.W.; Rhee, S.Y.; Ward, J.; Dickerson, J.; Lange, B.M.; Lane, G.; Roessner,
U.; Last, R. & Nikolau, B. (2007b). Minimum reporting standards for plant biology
context information in metabolomic studies. Metabolomics, Vol.3, No. 3, (Sep 2007),
pp. 195-201, issn 1573-3882
Fiehn, O.; Wohlgemuth, G.; Scholz, M.; Kind, T.; Lee, D.Y.; Lu, Y.; Moon, S. & Nikolau, B.
(2008). Quality control for plant metabolomics: reporting MSI-compliant studies.
Plant Journal, Vol.53, No. 4, (Feb 2008), pp. 691-704, issn 0960-7412
Figueiredo, L.F.D.; Sine, B.; Chantereau, J.; Mestres, C.; Fliedel, G.; Rami, J.F.; Glaszmann,
J.C.; Deu, M. & Courtois, B. (2010). Variability of grain quality in sorghum:
association with polymorphism in Sh2, Bt2, SssI, Ae1, Wx and O2. Theoretical and
Applied Genetics, Vol.121, No. 6, (Oct 2010), pp. 1171-1185, issn 0040-5752
Fouad, M.; Jabasini, M.; Kaji, N.; Terasaka, K.; Tokeshi, M.; Mizukami, H. & Baba, Y. (2008).
Microchip analysis of plant glucosinolates. Electrophoresis, Vol.29, No. 11, (Jun
2008), pp. 2280-2287, issn 0173-0835
Gaquerel, E.; Weinhold, A. & Baldwin, I.T. (2009). Molecular interactions between the
specialist herbivore Manduca sexta (Lepidoptera, Sphigidae) and its natural host
Nicotiana attenuata. VIII. An unbiased GCxGC-ToFMS analysis of the plant's elicited
volatile emissions. Plant Physiology, Vol.149, No. 3, (Mar 2009), pp. 1408-1423, issn
0032-0889
Giavalisco, P.; Kohl, K.; Hummel, J.; Seiwert, B. & Willmitzer, L. (2009). C-13 isotope-labeled
metabolomes allowing for improved compound annotation and relative
quantification in liquid chromatography-mass spectrometry-based metabolomic
research. Analytical Chemistry, Vol.81, No. 15, (Aug 2009), pp. 6546-6551, issn 0003-
2700
232 Metabolomics

Gibon, Y.; Vigeolas, H.; Tiessen, A.; Geigenberger, P. & Stitt, M. (2002). Sensitive and high
throughput metabolite assays for inorganic pyrophosphate, ADPGlc, nucleotide
phosphates, and glycolytic intermediates based on a novel enzymic cycling system.
Plant Journal, Vol.30, No. 2, (Apr 2002), pp. 221-235, issn 0960-7412
Hall, R.D. (2006). Plant metabolomics: from holistic hope, to hype, to hot topic. New
Phytologist, Vol.169, No. 3, (Jan 2006), pp. 453-468, issn 1469-8137
Hall, R.D. (2011a). Biology of Plant Metabolomics, In: Annual Plant Reviews, 420, Wiley-
Blackwell, isbn 978-1-4051-9954-4, Chichester, UK
Hall, R.D. (2011b). Plant Metabolomics in a Nutshell: Potential and Future Challenges, In:
Biology of Plant Metabolomics, Hall, R.D., (Ed.) 1-24, Wiley-Blackwell, isbn 978-1-
4051-9954-4, Chichester, UK
Hall, R.D.; Brouwer, I.D. & Fitzgerald, M.A. (2008). Plant metabolomics and its potential
application for human nutrition. Physiologia Plantarum, Vol.132, No. 2, (Feb 2008),
pp. 162-175, issn 0031-9317
Hall, R.D.; Wishart, D. & Roessner, U. (2011). Metabolomics and the move towards biology.
Metabolomics, Vol.7, No. 3, (Sep 2011), pp. 454-456, issn 1573-3882
Halpin, S.T. & Spence, D.M. (2010). Direct plate-reader measurement of nitric oxide released
from hypoxic erythrocytes flowing through a microfluidic device. Analytical
Chemistry, Vol.82, No. 17, (Sep 2010), pp. 7492-7497, issn 0003-2700
Hannemann, J.; Poorter, H.; Usadel, B.; Blasing, O.E.; Finck, A.; Tardieu, F.; Atkin, O.K.;
Pons, T.; Stitt, M. & Gibon, Y. (2009). Xeml Lab: a tool that supports the design of
experiments at a graphical interface and generates computer-readable metadata
files, which capture information about genotypes, growth conditions,
environmental perturbations and sampling strategy. Plant Cell and Environment,
Vol.32, No. 9, (Sep 2009), pp. 1185-1200, issn 0140-7791
Hausler, R.E.; Fischer, K.L. & Flugge, U.I. (2000). Determination of low-abundant
metabolites in plant extracts by NAD(P)H fluorescence with a microtiter plate
reader. Analytical Biochemistry, Vol.281, No. 1, (May 2000), pp. 1-8, issn 0003-2697
Hirai, M.Y.; Sawada, Y.; Kanaya, S.; Kuromori, T.; Kobayashi, M.; Klausnitzer, R.; Hanada,
K.; Akiyama, K.; Sakurai, T.; Saito, K. & Shinozaki, K. (2010). Toward genome-wide
metabolotyping and elucidation of metabolic system: metabolic profiling of large-
scale bioresources. Journal of Plant Research, Vol.123, No. 3, (May 2010), pp. 291-298,
issn 0918-9440
Hompesch, R.W.; Garcia, C.D.; Weiss, D.J.; Vivanco, J.M. & Henry, C.S. (2005). Analysis of
natural flavonoids by microchip-micellar electrokinetic chromatography with
pulsed amperometric detection. Analyst, Vol.130, No. 5, (May 2005), pp. 694-700,
issn 0003-2654
Jacob, S.S.; Smith, N.W. & Legido-Quigley, C. (2007). Assessment of Chinese medicinal herb
metabolite profiles by UPLC-MS-based methodology for detection of aristolochic
acids. Journal of Separation Science, Vol.30, No. 8, (May 2007), pp. 1200-1206, issn
1615-9306
Jemal, M.; Schuster, A. & Whigan, D.B. (2003). Liquid chromatography/tandem mass
spectrometry methods for quantitation of mevalonic acid in human plasma and
urine: method validation, demonstration of using a surrogate analyte, and
New Opportunities in Metabolomics and Biochemical Phenotyping for Plant Systems Biology 233

demonstration of unacceptable matrix effect in spite of use of a stable isotope


analog internal standard. Rapid Communications in Mass Spectrometry, Vol.17, No. 15,
(Jun 2003), pp. 1723-1734, issn 0951-4198
Jiang, G.F.; Attiya, S.; Ocvirk, G.; Lee, W.E. & Harrison, D.J. (2000). Red diode laser induced
fluorescence detection with a confocal microscope on a microchip for capillary
electrophoresis. Biosensors & Bioelectronics, Vol.14, No. 10-11, (Jan 2000), pp. 861-869,
issn 0956-5663
Joyce, A.R. & Palsson, B.O. (2006). The model organism as a system: integrating 'omics' data
sets. Nature Reviews Molecular Cell Biology, Vol.7, No. 3, (Mar 2006), pp. 198-210, issn
1471-0072
Kaiser, K.A.; Barding, G.A. & Larive, C.K. (2009). A comparison of metabolite extraction
strategies for 1H-NMR-based metabolic profiling using mature leaf tissue from the
model plant Arabidopsis thaliana. Magnetic Resonance in Chemistry, Vol.47, No. S1,
(Dec 2009), pp. S147-S156, issn 0749-1581
Keurentjes, J.J.B.; Angenent, G.C.; Dicke, M.; Dos Santos, V.; Molenaar, J.; van der Putten,
W.H.; de Ruiter, P.C.; Struik, P.C. & Thomma, B. (2011). Redefining plant systems
biology: from cell to ecosystem. Trends in Plant Science, Vol.16, No. 4, (Apr 2011),
pp. 183-190, issn 1360-1385
Keurentjes, J.J.B.; Fu, J.Y.; de Vos, C.H.R.; Lommen, A.; Hall, R.D.; Bino, R.J.; van der Plas,
L.H.W.; Jansen, R.C.; Vreugdenhil, D. & Koornneef, M. (2006). The genetics of plant
metabolism. Nature Genetics, Vol.38, No. 7, (Jul 2006), pp. 842-849, issn 1061-4036
Kim, H.K.; Choi, Y.H. & Verpoorte, R. (2011). NMR-based plant metabolomics: where do we
stand, where do we go? Trends in Biotechnology, Vol.29, No. 6, (Jun 2011), pp. 267-
275, issn 0167-7799
Kmellar, B.; Abranko, L.; Fodor, P. & Lehotay, S.J. (2010). Routine approach to qualitatively
screening 300 pesticides and quantification of those frequently detected in fruit and
vegetables using liquid chromatography tandem mass spectrometry (LC-MS/MS).
Food Additives and Contaminants Part a-Chemistry Analysis Control Exposure & Risk
Assessment, Vol.27, No. 10, (Oct 2010), pp. 1415-1430, issn 1944-0049
Koek, M.; Jellema, R.; van der Greef, J.; Tas, A. & Hankemeier, T. (2011). Quantitative
metabolomics based on gas chromatography mass spectrometry: status and
perspectives. Metabolomics, Vol.7, No. 3, (Sep 2011), pp. 307-328, issn 1573-3882
Kojima, M.; Kamada-Nobusada, T.; Komatsu, H.; Takei, K.; Kuroha, T.; Mizutani, M.;
Ashikari, M.; Ueguchi-Tanaka, M.; Matsuoka, M.; Suzuki, K. & Sakakibara, H.
(2009). Highly sensitive and high-throughput analysis of plant hormones using MS-
probe modification and liquid chromatography tandem mass spectrometry: An
application for hormone profiling in Oryza sativa. Plant and Cell Physiology, Vol.50,
No. 7, (Jul 2009), pp. 1201-1214, issn 0032-0781
Kozak, M.; Bocianowski, J.; Liersch, A.; Tartanus, M.g.; Bartkowiak-Broda, I.; Piotto, F. &
Azevedo, R. (2011). Genetic divergence is not the same as phenotypic divergence.
Molecular Breeding, Vol.28, No. 2, (May 2011), pp. 277-280, issn 1380-3743
Krueger, S.; Giavalisco, P.; Krall, L.; Steinhauser, M.C.; Bussis, D.; Usadel, B.; Flugge, U.I.;
Fernie, A.R.; Willmitzer, L. & Steinhauser, D. (2011). A topological map of the
234 Metabolomics

compartmentalized Arabidopsis thaliana leaf metabolome. Plos One, Vol.6, No. 3,


(Mar 2011), pp. 16, issn 1932-6203
Kusano, M.; Fukushima, A.; Redestig, H. & Saito, K. (2011a). Metabolomic approaches
toward understanding nitrogen metabolism in plants. Journal of Experimental
Botany, Vol.62, No. 4, (Feb 2011a), pp. 1439-1453, issn 0022-0957
Kusano, M.; Redestig, H.; Hirai, T.; Oikawa, A.; Matsuda, F.; Fukushima, A.; Arita, M.;
Watanabe, S.; Yano, M.; Hiwasa-Tanase, K.; Ezura, H. & Saito, K. (2011b). Covering
chemical diversity of genetically-modified tomatoes using metabolomics for
objective substantial equivalence assessment. Plos One, Vol.6, No. 2, (Feb 2011b),
issn 1932-6203
Lei, Z.; Huhman, D. & Sumner, L.W. (2011). Mass spectrometry strategies in metabolomics.
Journal of Biological Chemistry, (Jun 2011), issn 0021-9258
Leiss, K.A.; Choi, Y.H.; Verpoorte, R. & Klinkhamer, P.G.L. (2010). An overview of NMR-
based metabolomics to identify secondary plant compounds involved in host plant
resistance. Phytochemistry Reviews, Vol.10, No. 2, (Jun 2010), pp. 205-216, issn 1568-
7767
Lexa, M.; Genkov, T.; Malbeck, J.; Machackova, I. & Brzobohaty, B. (2003). Dynamics of
endogenous cytokinin pools in tobacco seedlings: A modelling approach. Annals of
Botany, Vol.91, No. 5, (Apr 2003), pp. 585-597, issn 0305-7364
Liu, G.N.; Zhu, Y.H. & Jiang, J.G. (2009). The metabolomics of carotenoids in engineered cell
factory. Applied Microbiology and Biotechnology, Vol.83, No. 6, (Jul 2009), pp. 989-999,
issn 0175-7598
Lu, W.; Bennett, B.D. & Rabinowitz, J.D. (2008). Analytical strategies for LC-MS-based
targeted metabolomics. Journal of Chromatography B-Analytical Technologies in the
Biomedical and Life Sciences, Vol.871, No. 2, (Aug 2008), pp. 236-242, issn 1570-0232
McDonald, J.C.; Duffy, D.C.; Anderson, J.R.; Chiu, D.T.; Wu, H.K.; Schueller, O.J.A. &
Whitesides, G.M. (2000). Fabrication of microfluidic systems in
poly(dimethylsiloxane). Electrophoresis, Vol.21, No. 1, (Jan 2000), pp. 27-40, issn
0173-0835
McDougall, G.; Martinussen, I. & Stewart, D. (2008). Towards fruitful metabolomics: High
throughput analyses of polyphenol composition in berries using direct infusion
mass spectrometry. Journal of Chromatography B-Analytical Technologies in the
Biomedical and Life Sciences, Vol.871, No. 2, (Aug 2008), pp. 362-369, issn 1570-0232
Meyer, R.C.; Steinfath, M.; Lisec, J.; Becher, M.; Witucka-Wall, H.; Torjek, O.; Fiehn, O.;
Eckardt, A.; Willmitzer, L.; Selbig, J. & Altmann, T. (2007). The metabolic signature
related to high plant growth rate in Arabidopsis thaliana. Proceedings of the National
Academy of Sciences of the United States of America, Vol.104, No. 11, (Mar 2007), pp.
4759-4764, issn 0027-8424
Mittler, R. & Blumwald, E. (2010). Genetic engineering for modern agriculture: challenges
and perspectives, In: Annual Review of Plant Biology, Merchant, S., Briggs, W.R., Ort,
D., (Eds) 443-462, isbn 978-0-8243-0661-8, Palo Alto, California
Moco, S.; Bino, R.J.; De Vos, R.C.H. & Vervoort, J. (2007). Metabolomics technologies and
metabolite identification. Trends in Analytical Chemistry, Vol.26, No. 9, (Oct 2007),
issn 01659936
New Opportunities in Metabolomics and Biochemical Phenotyping for Plant Systems Biology 235

Moco, S.; Schneider, B. & Vervoort, J. (2009). Plant micrometabolomics: The analysis of
endogenous metabolites present in a plant cell or tissue. Journal of Proteome
Research, Vol.8, No. 4, (Apr 2009), pp. 1694-1703, issn 1535-3893
Moing, A.; Aharoni, A.; Biais, B.; Rogachev, I.; Meir, S.; Brodsky, L.; Allwood, J.W.; Erban,
A.; Dunn, W.B.; Kay, L.; de Koning, S.; de Vos, R.C.H.; Jonker, H.; Mumm, R.;
Deborde, C.; Maucourt, M.; Bernillon, S.; Gibon, Y.; Hansen, T.H.; Husted, S.;
Goodacre, R.; Kopka, J.; Schjoerring, J.K.; Rolin, D. & Hall, R.D. (2011). Extensive
metabolic cross-talk in melon fruit revealed by spatial and developmental
combinatorial metabolomics. New Phytologist, Vol.190, No. 3, (May 2011), pp. 683-
696, issn 1469-8137
Moser, I.; Jobst, G. & Urban, G.A. (2002). Biosensor arrays for simultaneous measurement of
glucose, lactate, glutamate, and glutamine. Biosensors & Bioelectronics, Vol.17, No. 4,
(Apr 2002), pp. 297-302, issn 0956-5663
Mounet, F.; Moing, A.; Garcia, V.; Petit, J.; Maucourt, M.; Deborde, C.; Bernillon, S.; Le Gall,
G.; Colquhoun, I.; Defernez, M.; Giraudel, J.-L.; Rolin, D.; Rothan, C. & Lemaire-
Chamley, M. (2009). Gene and metabolite regulatory network analysis of early
developing fruit tissues highlights new candidate genes for the control of tomato
fruit composition and development. Plant Physiology, Vol.149, No. 3, (March 2009),
pp. 1505-1528, issn 0032-0889
Mulas, G.; Galaffu, M.G.; Pretti, L.; Nieddu, G.; Mercenaro, L.; Tonelli, R. & Anedda, R.
(2011). NMR analysis of seven selections of Vermentino grape berry: metabolites
composition and development. Journal of Agricultural and Food Chemistry, Vol.59,
No. 3, (Feb 2011), pp. 793-802, issn 0021-8561
Myles, S.; Peiffer, J.; Brown, P.J.; Ersoz, E.S.; Zhang, Z.W.; Costich, D.E. & Buckler, E.S.
(2009). Association mapping: Critical considerations shift from genotyping to
experimental design. Plant Cell, Vol.21, No. 8, (Aug 2009), pp. 2194-2202, issn 1040-
4651
Nagele, T.; Kandel, B.A.; Frana, S.; Meissner, M. & Heyer, A.G. (2011). A systems biology
approach for the analysis of carbohydrate dynamics during acclimation to low
temperature in Arabidopsis thaliana. Febs Journal, Vol.278, No. 3, (Feb 2011), pp. 506-
518, issn 1742-464X
Nagrath, S.; Sequist, L.V.; Maheswaran, S.; Bell, D.W.; Irimia, D.; Ulkus, L.; Smith, M.R.;
Kwak, E.L.; Digumarthy, S.; Muzikansky, A.; Ryan, P.; Balis, U.J.; Tompkins, R.G.;
Haber, D.A. & Toner, M. (2007). Isolation of rare circulating tumour cells in cancer
patients by microchip technology. Nature, Vol.450, No. 7173, (Dec 2007), pp. 1235-
1239, issn 0028-0836
Oikawa, A.; Matsuda, F.; Kusano, M.; Okazaki, Y. & Saito, K. (2008). Rice metabolomics.
Rice, Vol.1, No. 1, (Sep 2008), pp. 63-71, issn 1939-8425
Okada, T.; Afendi, F.M.; Altaf-Ul-Amin, M.; Takahashi, H.; Nakamura, K. & Kanaya, S.
(2010). Metabolomics of medicinal plants: The importance of multivariate analysis
of analytical chemistry data. Current Computer-Aided Drug Design, Vol.6, No. 3, (Sep
2010), pp. 179-196, issn 1573-4099
236 Metabolomics

Oliver, S.G.; Winson, M.K.; Kell, D.B. & Baganz, F. (1998). Systematic functional analysis of
the yeast genome. Trends in Biotechnology, Vol.16, No. 9, (Sep 1998), pp. 373-378,
issn 0167-7799
Palama, T.L.; Khatib, A.; Choi, Y.H.; Payet, B.; Fock, I.; Verpoorte, R. & Kodja, H. (2009).
Metabolic changes in different developmental stages of Vanilla planifolia pods.
Journal of Agricultural and Food Chemistry, Vol.57, No. 17, (Sep 2009), pp. 7651-7658,
issn 0021-8561
Pichersky, E. & Lewinsohn, E. (2011). Convergent evolution in plant specialized metabolism,
In: Annual Review of Plant Biology, 549-566, Annual Reviews, isbn 978-0-8243-0662-
5, Palo Alto, California
Poorter, H.; Niinemets, U.; Walter, A.; Fiorani, F. & Schurr, U. (2010). A method to construct
dose-response curves for a wide range of environmental factors and plant traits by
means of a meta-analysis of phenotypic data. Journal of Experimental Botany, Vol.61,
No. 8, (May 2010), pp. 2043-2055, issn 0022-0957
Poorter, H.; Walter, A.; Fiorani, F.; Schurr, U. & Niinemets, U. (2009). Meta-phenomics:
Building a unified framework for interpreting plant growth responses to diverse
environmental variables. Comparative Biochemistry and Physiology a-Molecular &
Integrative Physiology, Vol.153A, No. 2, (Jun 2009), pp. S224-S224, issn 1095-6433
Ramautar, R.; Mayboroda, O.A.; Somsen, G.W. & de Jong, G.J. (2011). CE-MS for
metabolomics: Developments and applications in the period 2008-2010.
Electrophoresis, Vol.32, No. 1, (Jan 2011), pp. 52-65, issn 0173-0835
Rashed, M.S.; Bucknall, M.P.; Little, D.; Awad, A.; Jacob, M.; Alamoudi, M.; Alwattar, M. &
Ozand, P.T. (1997). Screening blood spots for inborn errors of metabolism by
electrospray tandem mass spectrometry with a microplate batch process and a
computer algorithm for automated flagging of abnormal profiles. Clinical Chemistry,
Vol.43, No. 7, (Jul 1997), pp. 1129-1141, issn 0009-9147
Redestig, H. & Costa, I.G. (2011). Detection and interpretation of metabolite-transcript
coresponses using combined profiling data. Bioinformatics, Vol.27, No. 13, (Jul 2011),
pp. I357-I365, issn 1367-4803
Redestig, H.; Kusano, M.; Fukushima, A.; Matsuda, F.; Saito, K. & Arita, M. (2010).
Consolidating metabolite identifiers to enable contextual and multi-platform
metabolomics data analysis. BMC Bioinformatics, Vol.11, (Apr 2010), issn 1471-2105
Roda, A.; Pasini, P.; Mirasoli, M.; Michelini, E. & Guardigli, M. (2004). Biotechnological
applications of bioluminescence and chemiluminescence. Trends in Biotechnology,
Vol.22, No. 6, (Jun 2004), pp. 295-303, issn 0167-7799
Ruane, J. & Sonnino, A. (2011). Agricultural biotechnologies in developing countries and
their possible contribution to food security. Journal of Biotechnology, Vol.In Press,
Corrected Proof, ( 2011), issn 0168-1656
Ryan, D. & Robards, K. (2006). Analytical chemistry considerations in plant metabolomics.
Separation and Purification Reviews, Vol.35, No. 4, (Nov 2006), pp. 319-356, issn 1542-
2119
Saito, K. & Matsuda, F. (2010). Metabolomics for Functional Genomics, Systems Biology, and
Biotechnology, In: Annual Review of Plant Biology, Vol 61, 463-489, Annual Reviews,
isbn 1543-5008, Palo Alto, California
New Opportunities in Metabolomics and Biochemical Phenotyping for Plant Systems Biology 237

Salse, J. (2004). New in silico insight into the synteny between rice (Oryza sativa L.) and
maize (Zea mays L.) highlights reshuffling and identifies new duplications in the
rice genome (Vol.38, (May 2004), pp. 396-409). Plant Journal, Vol.38, No. 5, (Jun
2004), pp. 873-873, issn 0960-7412
Sanchez-Perez, E.M.; Iglesias, M.J.; Lopez-Ortiz, F.; Sanchez-Perez, I. & Martinez-Galera, M.
(2010). Study of the suitability of HRMAS NMR for metabolic profiling of tomatoes:
Application to tissue differentiation and fruit ripening. Food Chemistry, Vol.122, No.
3, (Oct 2010), pp. 877-887, issn 0308-8146
Sanchez, D.H.; Pieckenstain, F.L.; Szymanski, J.; Erban, A.; Bromke, M.; Hannah, M.A.;
Kraemer, U.; Kopka, J. & Udvardi, M.K. (2011). Comparative functional genomics
of salt stress in related model and cultivated plants identifies and overcomes
limitations to translational genomics. Plos One, Vol.6, No. 2, (Feb 2011), issn 1932-
6203
Sawada, Y.; Akiyama, K.; Sakata, A.; Kuwahara, A.; Otsuki, H.; Sakurai, T.; Saito, K. & Hirai,
M.Y. (2009). Widely targeted metabolomics based on large-scale ms/ms data for
elucidating metabolite accumulation patterns in plants. Plant and Cell Physiology,
Vol.50, No. 1, (Jan 2009), pp. 37-47, issn 0032-0781
Schauer, N.; Semel, Y.; Roessner, U.; Gur, A.; Balbo, I.; Carrari, F.; Pleban, T.; Perez-Melis, A.;
Bruedigam, C.; Kopka, J.; Willmitzer, L.; Zamir, D. & Fernie, A.R. (2006).
Comprehensive metabolic profiling and phenotyping of interspecific introgression
lines for tomato improvement. Nature Biotechnology, Vol.24, No. 4, (Apr 2006), pp.
447-454, issn 1087-0156
Shaw, R.A.; Rigatto, C.; Reslerova, M.; Ying, S.L.; Man, A.; Schattka, B.; Battrell, C.F.;
Matthewson, J. & Mansfield, C. (2009). Toward point-of-care diagnostic metabolic
fingerprinting: quantification of plasma creatinine by infrared spectroscopy of
microfluidic-preprocessed samples. Analyst, Vol.134, No. 6, (Jun 2009), pp. 1224-
1231, issn 0003-2654
Shepherd, L.V.T.; Fraser, P. & Stewart, D. (2011). Metabolomics: a second-generation
platform for crop and food analysis. Bioanalysis, Vol.3, No. 10, (May 2011), pp. 1143-
1159, issn 1757-6180
Shintu, L.; Le Gall, G. & Colquhoun, I.J. (2009). Metabolomics and the detection of
unintended effects in genetically modified crops, In: Plant-derived Natural Products,
Osbourn, A.E., Lanzotti, V., (Eds) 505-531, Springer New York, isbn 978-0-387-
85497-7, New-York
Shroff, R.; Rulisek, L.; Doubsky, J. & Svatos, A. (2009). Acid-base-driven matrix-assisted
mass spectrometry for targeted metabolomics. Proceedings of the National Academy of
Sciences of the United States of America, Vol.106, No. 25, (Jun 2009), pp. 10092-10096,
issn 0027-8424
Skogerson, K.; Harrigan, G.G.; Reynolds, T.L.; Halls, S.C.; Ruebelt, M.; Iandolino, A.;
Pandravada, A.; Glenn, K.C. & Fiehn, O. (2010). Impact of genetics and
environment on the metabolite composition of maize grain. Journal of Agricultural
and Food Chemistry, Vol.58, No. 6, (Mar 2010), pp. 3600-3610, issn 0021-8561
238 Metabolomics

Smith, J.E. & Bluhm, B.H. (2011). Metabolic fingerprinting in Fusarium verticillioides to
determine gene function, In: Fungal Genomics : Methods and Protocols, Xu, J.R.,
Bluhm, B.H.H., (Eds) 237-247, Humana Press, isbn 978-1-61779-039-3, Heidelberg
Stitt, M.; Gibon, Y.; Lunn, J.E. & Piques, M. (2007). Multilevel genomics analysis of carbon
signalling during low carbon availability: coordinating the supply and utilisation of
carbon in a fluctuating environment. Functional Plant Biology, Vol.34, No. 6, (Jun
2007), pp. 526-549, issn 1445-4408
Sulpice, R.; Trenkamp, S.; Steinfath, M.; Usadel, B.; Gibon, Y.; Witucka-Wall, H.; Pyl, E.T.;
Tschoep, H.; Steinhauser, M.C.; Guenther, M.; Hoehne, M.; Rohwer, J.M.; Altmann,
T.; Fernie, A.R. & Stitt, M. (2010). Network analysis of enzyme activities and
metabolite levels and their relationship to biomass in a large panel of Arabidopsis
accessions. Plant Cell, Vol.22, No. 8, (Aug 2010), pp. 2872-2893, issn 1040-4651
Sumner, L. (2010). Recent advances in plant metabolomics and greener pastures. F1000
Biology Reports, Vol.2 (Jan 2010), pp. 7, issn 1757-594X
Takatsy, G. (1955). The use of spiral loops in serological and virological micro-methods. Acta
Microbiologica Academiae Scientiarum Hungaricae, Vol. 3, No. 1-2, (Jun 1955), pp. 191-
202, issn 0001-6187
Tarpley, L.; Duran, A.L.; Kebrom, T.H. & Sumner, L.W. (2005). Biomarker metabolites
capturing the metabolite variance present in a rice plant developmental period.
BMC Plant Biology, Vol.5, (May 2005), issn 1471-2229
Tester, M. & Langridge, P. (2010). Breeding technologies to increase crop production in a
changing world. Science, Vol.327, No. 5967, (Feb 2010), pp. 818-822, issn 0036-8075
Thiele, B.; Fullner, K.; Stein, N.; Oldiges, M.; Kuhn, A.J. & Hofmann, D. (2008). Analysis of
amino acids without derivatization in barley extracts by LC-MS-MS. Analytical and
Bioanalytical Chemistry, Vol.391, No. 7, (Aug 2008), pp. 2663-2672, issn 1618-2642
Tikunov, Y.M.; Verstappen, F.W.A. & Hall, R.D. (2007). Metabolomic profiling of natural
volatiles headspace trapping: GC-MS, In: Metabolomics, Weckwerth, W., (Ed.) 39-53,
Humana Press, isbn 1064-3745, Totowa, New Jersey
Tomas, R.; Kleparnik, K. & Foret, F. (2008). Multidimensional liquid phase separations for
mass spectrometry. Journal of Separation Science, Vol.31, No. 11, (Jun 2008), pp. 1964-
1979, issn 1615-9306
Trethewey, R.N. (2004). Metabolite profiling as an aid to metabolic engineering in plants.
Current Opinion in Plant Biology, Vol.7, No. 2, (Apr 2004), pp. 196-201, issn 1369-5266
Trufelli, H.; Palma, P.; Famiglini, G. & Cappiello, A. (2011). An overview of matrix effects in
liquid chromatography-mass spectrometry. Mass Spectrometry Reviews, Vol.30, No.
3, (May-Jun 2011), pp. 491-509, issn 0277-7037
Urbanczyk-Wochniak, E.; Baxter, C.; Kolbe, A.; Kopka, J.; Sweetlove, L.J. & Fernie, A.R.
(2005). Profiling of diurnal patterns of metabolite and transcript abundance in
potato (Solanum tuberosum) leaves. Planta, Vol.221, No. 6, (Aug 2005), pp. 891-903,
issn 0032-0935
Usadel, B.; Blasing, O.E.; Gibon, Y.; Retzlaff, K.; Hoehne, M.; Gunther, M. & Stitt, M. (2008).
Global transcript levels respond to small changes of the carbon status during
progressive exhaustion of carbohydrates in Arabidopsis rosettes. Plant Physiology,
Vol.146, No. 4, (Apr 2008), pp. 1834-1861, issn 0032-0889
New Opportunities in Metabolomics and Biochemical Phenotyping for Plant Systems Biology 239

Vengasandra, S.; Cai, Y.K.; Grewell, D.; Shinar, J. & Shinar, R. (2010). Polypropylene CD-
organic light-emitting diode biosensing platform. Lab on a Chip, Vol.10, No. 8, 2010),
pp. 1051-1056, issn 1473-0197
Vidal, M. (2009). A unifying view of 21st century systems biology. Febs Letters, Vol.583, No.
24, (Dec 2009), pp. 3891-3894, issn 0014-5793
Villiers, F.; Ducruix, C.; Hugouvieux, V.; Jarno, N.; Ezan, E.; Garin, J.; Junot, C. &
Bourguignon, J. (2011). Investigating the plant response to cadmium exposure by
proteomic and metabolomic approaches. Proteomics, Vol.11, No. 9, (May 2011), pp.
1650-1663, issn 1615-9861
Wang, Q.Z.; Wu, C.Y.; Chen, T.; Chen, X. & Zhao, X.M. (2006). Integrating metabolomics
into a systems biology framework to exploit metabolic complexity: strategies and
applications in microorganisms. Applied Microbiology and Biotechnology, Vol.70, No.
2, (Mar 2006), pp. 151-161, issn 0175-7598
Wang, X.Y.; Gowik, U.; Tang, H.B.; Bowers, J.E.; Westhoff, P. & Paterson, A.H. (2009).
Comparative genomic analysis of C4 photosynthetic pathway evolution in grasses.
Genome Biology, Vol.10, No. 6, (Jun 2009), issn 1474-760X
Ward, J.; Baker, J.M.; Miller, S.; Deborde, C.; Maucourt, M.; Biais, B.; Rolin, D.; Moing, A.;
Moco, S.; Vervoort, J.; Lommen, A.; Schäfer, H.; Humpfer, E. & Beale, M.H. (2010).
An inter-laboratory comparison demonstrates that [1H]-NMR metabolite
fingerprinting is a robust technique for collaborative plant metabolomic data
collection. Metabolomics, Vol.6, No. 2, (Jun 2010), pp. 263-273, issn 1573-3882
Weckwerth, W. (2007). Metabolomics. Methods and protocols. Humana Press, isbn 1-59745-244-
0, Totowa, USA
Weckwerth, W. (2011). Unpredictability of metabolism-the key role of metabolomics science
in combination with next-generation genome sequencing. Analytical and
Bioanalytical Chemistry, Vol.400, No. 7, (Jun 2011), pp. 1967-1978, issn 1618-2642
Weckwerth, W.; Loureiro, M.E.; Wenzel, K. & Fiehn, O. (2004). Differential metabolic
networks unravel the effects of silent plant phenotypes. Proceedings of the National
Academy of Sciences of the United States of America, Vol.101, No. 20, (May 2004), pp.
7809-7814, issn 0027-8424
Werner, E.; Heilier, J.F.; Ducruix, C.; Ezan, E.; Junot, C. & Tabet, J.C. (2008). Mass
spectrometry for the identification of the discriminating signals from
metabolomics: Current status and future trends. Journal of Chromatography B-
Analytical Technologies in the Biomedical and Life Sciences, Vol.871, No. 2, (Aug 2008),
pp. 143-163, issn 1570-0232
Westerhoff, H.V.; Winder, C.; Messiha, H.; Simeonidis, E.; Adamczyk, M.; Verma, M.;
Bruggeman, F.J. & Dunn, W. (2009). Systems Biology: The elements and principles
of Life. Febs Letters, Vol.583, No. 24, (Dec 2009), pp. 3882-3890, issn 0014-5793
Whitesides, G.M. (2006). The origins and the future of microfluidics. Nature, Vol.442, No.
7101, (Jul 2006), pp. 368-373, issn 0028-0836
Williams, T.C.R.; Poolman, M.G.; Howden, A.J.M.; Schwarzlander, M.; Fell, D.A.; Ratcliffe,
R.G. & Sweetlove, L.J. (2010). A genome-scale metabolic model accurately predicts
fluxes in central carbon metabolism under stress conditions. Plant Physiology,
Vol.154, No. 1, (Sep 2010), pp. 311-323, issn 0032-0889
240 Metabolomics

Wishart, D.S. (2008). Metabolomics: applications to food science and nutrition research.
Trends in Food Science & Technology, Vol.19, No. 9, 2008), pp. 482-493, issn 0924-2244
Wurm, M.; Schopke, B.; Lutz, D.; Muller, J. & Zeng, A.P. (2010). Microtechnology meets
systems biology: The small molecules of metabolome as next big targets. Journal of
Biotechnology, Vol.149, No. 1-2, (Aug 2010), pp. 33-51, issn 0168-1656
Yu, J.M.; Holland, J.B.; McMullen, M.D. & Buckler, E.S. (2008). Genetic design and statistical
power of nested association mapping in maize. Genetics, Vol.178, No. 1, (Jan 2008),
pp. 539-551, issn 0016-6731
Zhang, N.Y.; Gur, A.; Gibon, Y.; Sulpice, R.; Flint-Garcia, S.; McMullen, M.D.; Stitt, M. &
Buckler, E.S. (2010). Genetic analysis of central carbon metabolism unveils an
amino acid substitution that alters maize NAD-dependent isocitrate
dehydrogenase activity. Plos One, Vol.5, No. 3, (Apr 2010), issn 1932-6203
Zhang, Y.; Thiele, I.; Weekes, D.; Li, Z.W.; Jaroszewski, L.; Ginalski, K.; Deacon, A.M.;
Wooley, J.; Lesley, S.A.; Wilson, I.A.; Palsson, B.; Osterman, A. & Godzik, A. (2009).
Three-dimensional structural view of the central metabolic network of Thermotoga
maritima. Science, Vol.325, No. 5947, (Sep 2009), pp. 1544-1549, issn 0036-8075
10

Metabolomics of Endophytic Fungi Producing


Associated Plant Secondary Metabolites:
Progress, Challenges and Opportunities
Souvik Kusari and Michael Spiteller
Institute of Environmental Research (INFU) of the Faculty of Chemistry, Chair of
Environmental Chemistry and Analytical Chemistry, TU Dortmund, Dortmund,
Germany

1. Introduction
Microorganisms are indispensable for every aspect of human life, in fact all life on earth,
although they cannot be seen by the naked eye. Since time immemorial, every process in the
biosphere has been affected by the apparently unending ability of microbes to renovate the
world around them. More recently, many discoveries have been made in isolating a special
class of microorganisms, mainly fungi but also bacteria, commonly called endophytes,
which have been shown to have the natural potential for accumulation of various bioactive
metabolites which may directly or indirectly be used as therapeutic agents against a
plethora of maladies (Kusari & Spiteller, 2010, 2011). Bioprospecting endophytes have led to
exciting possibilities to explore and utilize their potential. Several bioprospecting strategies
might be employed in order to discover potent endophytes with desirable traits (Figure 1).
These include randomly sampling different plants from any population to isolate the
associated endophytes, or first performing a detailed study of an ecosystem in order to
determine its features with regard to its natural population of plant species, their
relationship with the environment, soil composition, and biogeochemical cycles. Another
approach is to evaluate the evolutionary relatedness among groups of plants at a particular
sampling site, correlating to species, genus, and populations, through morphological data
matrices and molecular sequencing, followed by isolation of endophytes from the desired
plants. Traditional medicinal plants are also bioprospected for endophytes, especially for the
ones capable of producing one or more of the bioactive secondary metabolites present in the
host plants. Finally, the valuable information obtained using the different bioprospecting
schemes can be pooled together, comparatively evaluated, and stored for further use
applying suitable data mining approaches.

2. What is an endophytic fungus?


Endophytes are microorganisms that internally infect living plant tissues without causing
any visible manifestation of disease, and live in mutualistic association with plants for at
least a part of their life cycle (Bacon & White, 2000). The term ‘endophyte’ (Gr. endon, within;
phyton, plant) was first contrived by de Bary (1866). All types of microorganisms (fungi,
242 Metabolomics

Fig. 1. Different bioprospecting strategies that might be utilized in order to discover novel or
competent endophytes with desirable features.

bacteria, and actinomycetes) have been discovered as endophytes. The most frequently
encountered endophytes are fungi (Staniek et al., 2008). Fungal endophytes constitute an
inexplicably diverse group of polyphyletic fungi ubiquitous in plants, and maintain an
indiscernible dynamic relationship with their hosts for at least a part of their life cycle
(Figure 2a,b). The existence of fungi inside the tissues of asymptomatic plants has been
known since the end of the nineteenth century (Guerin, 1898). Evidence of plant-associated
microorganisms found in the fossilized tissues of stems and leaves has revealed that
endophyte-plant associations may have evolved from the time higher plants first appeared
on the earth (Redecker et al., 2000). However, except for some infrequent studies, it was not
until the end of the twentieth century that fungal endophytes began to receive more
attention from scientists. Since endophytes were first described in the Darnel (Freeman,
1904), various investigators have isolated endophytes from different plant species. These
discoveries led to a worldwide search for novel endophytes for the better understanding
and applicability of such a promising group of microorganisms. On the one hand, the
ecological aspects of endophytic fungi such as host range, evolutionary relatedness,
infection, colonization, transmission patterns, tissue specificity, and mutualistic fitness
benefits have been investigated relating to a plethora of plants (Arnold et al., 2003, 2007,
Arnold, 2005, 2007; Stone et al., 2004; Schulz &Boyle, 2005; Rodriguez et al., 2009) (Figure
2c). On the other hand, many discoveries have been made in isolating endophytic fungi,
which have been shown to have the potential for de novo synthesis of various bioactive
metabolites that may directly or indirectly be used as therapeutic agents against numerous
ailments (Strobel and Daisy, 2003; Strobel et al., 2004; Zhang et al., 2006; Gunatilaka, 2006;
Staniek et al., 2008; Suryanarayanana et al., 2009; Aly et al., 2010; Kharwar et al., 2011; Kusari
& Spiteller, 2010, 2011).
Metabolomics of Endophytic Fungi Producing
Associated Plant Secondary Metabolites: Progress, Challenges and Opportunities 243

Fig. 2. A diagrammatic representation of endophytic fungal association with their host


plants. (a) Possible life histories of endophytic fungi pre- and post-‘endophytism’.
(b) Different localization patterns of fungal endophytes within plant tissues. (c) Location of
the different classes of endophytes (according to Rodriguez et al., 2009). *An artistic
rendition of the possible thoughts of a plant about its endophytic counterparts, presented in
memory of Cole Albert Porter (1891-1964) and Frank Sinatra (1915-1998).
244 Metabolomics

3. Endophytic fungi producing host plant secondary metabolites


The possibility that endophytes biosynthesize associated plant compounds was first
comprehended and published by Stierle et al. (1993), following the highly heralded
discovery of endophytic Taxomyces andreanae that produces the multi-billion dollar
anticancer compound Taxol® (generic name: paclitaxel), which was isolated from the Pacific
yew tree Taxus brevifolia. Inspired by this discovery, numerous efforts have been made to
identify endophytes as sources of associated plant natural products. Endophytes producing
antineoplastic camptothecin (CPT) and its structural analogs (Puri et al., 2005; Kusari et al.,
2009b, 2011b; Shweta et al., 2010), anticancer pro-drugs podophyllotoxin (Eyberger et al.,
2006; Puri et al., 2006) and deoxypodophyllotoxin (Kusari et al., 2009a), antidepressants
hypericin and emodin (Kusari et al., 2008, 2009c), and natural insecticides azadirachtin A
and B (Kusari et al., 2011c) are some of the interesting discoveries that followed. Novel
fungal endophytes capable of producing other associated plant secondary metabolites of
therapeutic importance, such as artemisinin, morphine, cannabinoids, and many more,
remain to be discovered and characterized.

4. Rationale for plant selection to provide the best opportunities for isolating
endophytic fungi producing associated plant natural products
Considering the enormous numbers and the diversity of plants, ingenious strategies should
be utilized to narrow the search for endophytes producing plant compounds. A specific
rationale for the collection of each plant for endophyte isolation could be proposed to
maximize possibility of discovering endophytes equipped with the capacity to produce
associated plant natural products. Several hypotheses governing this plant selection strategy
might be exploited.

4.1 Plants from inimitable ecological niche, especially those with an uncommon
morphology and possessing unusual strategies for subsistence
4.1.1 Case study: Hypericum perforatum
Plants from a distinct ecological niche or with unusual biology might also harbor potent
endophytes. A fine example of such a plant is Hypericum perforatum, which is commonly
called St. John’s wort (Wichtl, 1986) (Figure 3a). This plant is a pseudogamous,
facultatively apomictic, perennial medicinal plant that is native to Europe, West and
South Asia, North Africa, North America, and Australia (Hickey and
King, 1981; Wichtl, 1986). In general, Hypericum has always been a very important
medicinal plant occupying a significant place in ancient history. Pedanius Dioscorides, the
foremost ancient Greek herbalist, mentioned four species of Hypericum - uperikon, askuron,
androsaimon, and koris, which he recommended for sciatica, “when drunk with 2 heim of
hydromel (honey water)” (Gunther, 1959). H. perforatum has also been in use, at least from
the time of ancient Greece (Tammaro and Xepapadakis, 1986), as an antidepressant, in
healing of wounds and menstrual disorders, due to the presence of the-then unknown
bioactive compounds in the plant. This plant has also found historical use in India, China,
Egypt and many countries of Europe, where the tribal peoples have been burning this
plant to represent sun, light, vitality and strength (Hickey & King, 1981). We know now
that this plant produces the widely used antidepressant compound hypericin (Brockmann
Metabolomics of Endophytic Fungi Producing
Associated Plant Secondary Metabolites: Progress, Challenges and Opportunities 245

et al., 1950). Hypericin is a photodynamic compound (Kubin et al., 2005) which is


localized and probably also synthesized in the ‘dark glands’ (Onelli et al., 2002), which are
small specialized glandular structures dispersed over all above-ground parts of the plant
(flowers, capsules, leaves, stems) but not in the roots (Hölzl et al., 2003) (Figure 3b,c).
Therefore, using the rationale that a plant with such an uncommon biology (dark glands)
for protecting itself from the photodynamic effects of its own metabolite might also
contain endophytes that have been evolutionarily co-adapted to accumulate the same or
similar molecules, we undertook bioprospecting endophytic fungi from H. perforatum
sampled from various populations across Europe and the Himalayan region. This led to
the discovery of an endophytic fungus associated with this plant capable of producing
hypericin and emodin in axenic cultures (Kusari et al., 2008, 2009c).

Fig. 3. Hypericum perforatum as a suitable example of bioprospecting plants with an unusual


morphology, possessing unusual strategies for existence, and from unique environments. (a)
Wild H. perforatum growing at the Himalayan environments (Harwan, Jammu and Kashmir,
India) from where the hypericin- and emodin-producing endophyte was isolated.
(Photograph courtesy of M. Spiteller). (b) A representative leaf of H. perforatum where the
dark glands (arrows) can be seen. (c) A brightfield microscopic image of the leaf adaxial
surface where the dark glands (arrow) can be seen as black spots (using Leica S8 APO
Greenough stereo microscope, Leica Microsystems GmbH, Wetzlar, Germany; scale = 1 mm.
The image was captured using Leica EC3 digital camera and processed using the Leica
Application Suite LAS EZ ver. 1.6.0).
246 Metabolomics

4.2 Plants that have an ethno-botanical history that is associated with the specific
practices or applications of interest
4.2.1 Case study: Juniperus species
Juniperus plants (Figure 4) serve an excellent example to describe this rationale, which
contain the therapeutically important anticancer lignans podophyllotoxin and
deoxypodophyllotoxin (Hartwell et al., 1953). This species was in use as early as in the first
century A.D., when Gaius Plinius Secundus mentioned that the smaller species of Juniperus
could be used, among other things, to stop tumors or swelling (Imbert, 1998). The use of the
oil of Juniperus species (J. sabina, J. phoenicea and J. communis) for the treatment of ulcers,
carbuncles and leprosy was also mentioned by Dioscorides (Gunther, 1959). Generally, the
dried needles, called savin, or the derived oil was used. In 47 A.D., Scribonius Largus wrote
that savin oil was used to soften “hard female genital parts” (Sconocchia, 1983). Savin was
later also used to treat uterine carcinoma, venereal warts and polyps. Based on such
historical use by indigenous people, we recently isolated and characterized endophytic
fungi harbored in Juniperus plants sampled from the natural populations in Dortmund and
Haltern, Germany, and Jammu and Kashmir, India. This resulted in the discovery of a
deoxypodophyllotoxin-producing endophytic fungus harbored in J. communis (Kusari et al.,
2009a).

Fig. 4. Juniperus as an example of bioprospecting plants having an ethnobotanical history


associated with the specific practices or applications of interest. Some Juniperus species
growing at Rombergpark, Dortmund, Germany are shown. (Photographs courtesy of S.
Kusari and M. Spiteller).

4.3 Endemic or endangered plants


4.3.1 Case study I: Camptotheca acuminata
Endemic plants are frequently consorted with ecological peculiarities or typical locations
that are geographically distinct. Many of these plants are getting vulnerable owing to
their therapeutic, agricultural, environmental, and commercial value. These distinct plants
might also harbor a plethora of unique endophytes. One out of many examples is the
plant Camptotheca acuminata (Figure 5a). This plant grows in mainland China, and is
commonly called the ‘happy tree’, which is a direct translation of the Chinese word ‘Xi
Shu’. Camptotheca was first recorded in 1848 and scientifically described and named by
Decaisne (1873). The genus name Camptotheca is from the Greek Campto (meaning, bent or
curved) and theca (meaning, a case) referring to the anthers, which are bent inward. The
species name acuminata is derived from acuminate, which refers to the tips of leaves. This
Metabolomics of Endophytic Fungi Producing
Associated Plant Secondary Metabolites: Progress, Challenges and Opportunities 247

plant has been in use as traditional medicine in China for treatment of psoriasis, liver and
stomach ailments and the common cold (Sung et al., 1998). The present application of this
plant is on account of the fact that it contains substantial quantities of an important
antineoplastic drug, namely camptothecin (CPT). This plant is uprooted and harvested by
various sectors, including medical groups, pharmaceutical companies and scientists from
around the world, to isolate CPT for numerous purposes (Lorence & Nessler, 2004;
Sankar-Thomas, 2010). In addition to the difficulties of the practical total synthesis of this
natural compound, the unpredictable problems of nature such as erratic weather and
pests (Kusari et al., 2011d) have rendered this plant species vulnerable to extinction. As
such, in 2000 and again in 2006, C. acuminata was proposed for protection in the CITES
(Convention for International Trade in Endangered Species), World Conservation
Monitoring Centre, appendix II (Anonymous, 2000, 2006). This appendix lists species that
are not necessarily now threatened with extinction but that may become so unless trade is
closely controlled. There are, of course, some nurseries growing C. acuminata for
commercial purposes. These few nurseries, however, cannot meet the demand for CPT
production (Sankar-Thomas, 2010). Furthermore, the yields of CPT from field trees vary
widely and depend on factors that are difficult to control. For instance, plant diseases
such as leaf spot and root rot are some of the major fungal diseases that can limit the
cultivation of Camptotheca plants (Li et al., 2005) and diminish the production of CPT.
Cultivation of Camptotheca plants is limited to subtropical climates and it takes about ten
years for plants to produce a stable fruit yield (Li et al., 2005; Sankar-Thomas, 2010). The
combination of a high demand for CPT and its scarcity from natural plant sources has,
therefore, led to a different strategy of bioprospecting the endophytic fungi associated
with the C. acuminata as alternate sources of CPT and related metabolites (Kusari et al.,
2009b, 2011b).

Fig. 5. Camptotheca acuminata and Nothapodytes nimmoniana as fitting examples of


bioprospecting endemic or endangered plants. (a) C. acuminata growing at the Southwest
Forestry University campus, Kunming, Yunnan Province, China. (b) N. nimmoniana growing
at the Western Ghats, India. (Photographs courtesy of M. Spiteller).
248 Metabolomics

4.3.2 Case study II: Nothapodytes nimmoniana


Another plant containing CPT, Nothapodytes nimmoniana of Indian origin (Figure 5b) has also
been subjected to extensive harvesting resulting in vulnerability of the plant. Unlike C.
acuminata, there are no major commercial plantations of N. nimmoniana (Shaanker et al.,
2008). This has led to harvesting of the plants sampled directly from the wild populations in
India to meet the present and projected demands of CPT. In fact, it is estimated that in the
last decade alone, there has been at least 20% decline in the population of N. nimmoniana,
leading to the red-listing of this species (Kumar & Ved, 2000; Hombe Gowda et al., 2002).
Therefore, a need for the preservation of this endangered plant species as well as to ensure a
continuous supply of CPT has been felt. Endophyte research on this plant has yielded
notable results (Puri et al., 2005).

4.4 Plants growing in areas of abundant biodiversity


4.4.1 Case study: Azadirachta indica
Plants growing in areas where a multitude of biotic and abiotic factors play essential roles
and generating bioactive natural products might harbor diverse endophytic population.
Azadirachta indica A. Juss. (synonym Melia azadirachta), commonly known as the Indian neem
or Indian lilac (Butterworth, 1968) is well known in India and its neighboring countries for
more than 2000 years as one of the most versatile medicinal plants growing abundantly in
regions having high biodiversity of plants (Biswas et al., 2002). Traditionally, neem-based
formulations have been used to cure a variety of ailments such as fever, pain, leprosy, and
malaria in Ayurvedic and Unani medical treatments, but the most striking property of neem
tree reported to date is its insect-repellent property (Veitch et al., 2008). It is now established
that neem plants contain the natural product azadirachtin (i.e., azadirachtin A and structural
analogues) that distresses insects as an antifeedent and insect growth regulator (Lay et al.,
1993). Numerous natural bioactive compounds have been isolated and characterized from
different parts of the neem tree, and new natural products are being discovered every year.
Thus, it would seem that A. indica is a potential natural resource sheltering unique and
competent endophytes having a multitude of desirable traits. As such, neem plants have not
only been studied for their endophytic microflora concerning the composition, diversity,
and distribution of endophytic microbes within the plants (Rajagopal & Suryanarayanan,
2000; Mahesh et al., 2005; Verma et al., 2007, 2009), but also for endophytes producing
bioactive natural products (Li et al., 2007; Kharwar et al., 2009; Wu et al., 2008, 2009).
Recently, we isolated an endophytic fungus from this plant that is capable of producing
azadirachtin A and B under in vitro axenic conditions (Kusari et al., 2011c).

5. Metabolomics of endophytic fungi producing associated plant natural


products
5.1 Anticancer compounds
5.1.1 Case study I: Camptothecin (CPT) and structural analogues
Camptothecin (CPT), a pentacyclic quinoline alkaloid, is a potent antineoplastic agent,
which was first isolated from the wood of Camptotheca acuminata Decaisne (Nyssaceae), a
plant native to mainland China (Wall et al., 1966). CPT and its structural analogues (Figure
Metabolomics of Endophytic Fungi Producing
Associated Plant Secondary Metabolites: Progress, Challenges and Opportunities 249

6) have emerged as one of the most promising agents for cancer treatment owing to the
typical action mechanism involving DNA-Topoisomerase I, i.e., they cause DNA damage by
stabilizing a normally transient covalent complex between Topoisomerase I (Topo 1) and
DNA (Hsiang et al., 1985; Kusari et al., 2011a,d). CPT interacts with the Topo 1-DNA
complex, thereby forming a ternary complex that stabilizes the trans-esterification
intermediate (Hertzberg et al., 1990; Pommier et al., 1995). Thus, by stabilizing the cleavable
complex, CPT transforms the normally useful enzyme Topo 1 into an intracellular, cytotoxic
poison, and hence, CPT and structural analogues are called ‘topoisomerase poisons’ or
‘topoisomerase inhibitors’ (Lorence & Nessler, 2004).
Endophytic Entrophospora infrequens (Puri et al., 2005; Amna et al., 2006) and Neurospora
crassa (Rehman et al., 2008) isolated from N. nimmoniana were initially reported to produce
CPT. However, in both cases, there have been no further studies on how the fungi are able
to produce CPT and prevent self-toxicity from the intracellular accumulated CPT. Further,
no follow-up work on up-scaling the production of CPT has been performed, and there is no
published breakthrough in the commercial exploitation of these endophytic fungi as a
source of CPT.

Fig. 6. Camptothecin (CPT) and some important natural analogues found in plants.

Recently, we isolated an endophytic fungus, Fusarium solani, from the inner bark of
Camptotheca acuminata Decaisne, obtained from the Southwest Forestry University (SWFU)
campus, Kunming (Yunnan Province), People’s Republic of China (Figure 7a). This
250 Metabolomics

endophyte is capable of indigenously producing CPT, 9-methoxycamptothecin (9-MeO-


CPT), and 10-hydroxycamptothecin (10-OH-CPT) in submerged in vitro axenic culture
(Kusari et al., 2009b). We further investigated how this endophytic fungus, capable of
producing CPT, ensures self-resistance before being incapacitated by its own and the
associated plant’s CPT biosyntheses. We discovered its survival strategy by examining the
fungal Top1 (Topo 1) structure with emphasis on the CPT-binding and catalytic domains
(Kusari et al., 2011a). The typical amino acid residues Asn352, Glu356, Arg488, Gly503, and
Gly717 (numbered according to human Top1) were identified that ensure fungal resistance
towards intracellular CPT. The substitution Met370Thr is identical to that found in CPT-
resistant human cancer cells, but different from the host C. acuminata. This work denoted the
significance of resistance mechanisms employed by endophytes to bear toxic host
metabolites, and provided a deeper understanding of plant-microbe coevolution.

Fig. 7. (a) Endophytic Fusarium solani that produces CPT, 9-MeO-CPT and 10-OH-CPT. (b) A
cross-species CPT biosynthetic pathway where the endophyte utilizes indigenous enzymes
to biosynthesize CPT precursors (10-hydroxygeraniol, secologanin, and tryptamine), but
requires the host strictosidine synthase to complete the pathway. (c) Perforations on the
leaves N. nimmoniana plant (Western Ghats, India) (red arrows) are caused by feeding of
Chrysomelid leaf beetles. (Photographs courtesy of M. Spiteller).
Metabolomics of Endophytic Fungi Producing
Associated Plant Secondary Metabolites: Progress, Challenges and Opportunities 251

The discovery of this endophytic fungus that is capable of producing CPT led us to envisage
the possibility of using this organism to produce CPT under controlled fermentation
conditions in an economical, environment-friendly, and reproducible manner amenable to
industrial scale-up. Unfortunately, it was observed that a substantial decrease occurred in
the production of CPT and 9-MeO-CPT by this in vitro-cultured endophyte following
repeated subculturing (i.e., in successive subculture generations) (Kusari et al., 2009b).
Optimized fermentation conditions and the addition of precursors as well as various host
plant tissue extracts did not restore the production. We then deciphered the chemical
ecology of the endophyte-host interaction, where the fungal endophyte utilizes indigenous
G10H (geraniol 10-hydroxylase), SLS (secologanin synthase), and TDC (tryptophan
decarboxylase) to biosynthesize CPT precursors. However, to complete the cross-species
CPT biosynthetic pathway, the endophyte requires the host STR (strictosidine synthase)
(Kusari et al., 2011b) (Figure 7b). The fungal CPT biosynthetic genes destabilized ex planta
over successive subculture generations. The seventh subculture predicted proteins exhibited
reduced homologies to the original enzymes proving that such genomic instability leads to
dysfunction at the amino acid level. The endophyte with an impaired CPT biosynthetic
capability was artificially inoculated into the living host plants and then recovered after
colonization. CPT biosynthesis could still not be restored. This demonstrated that the
observed phenomenon of genomic instability was possibly irreversible (Kusari et al., 2011b).
Following our discovery of the endophytic fungus F. solani, another endophytic fungus has
been isolated from Apodytes dimidiata capable of producing the same compounds (Shweta et
al., 2010). Furthermore, an endophytic Xylaria sp. has recently been isolated from C.
acuminata capable of producing only 10-OH-CPT, and remarkably, not the parent compound
CPT (Liu et al., 2010). In both these cases, no further follow-up studies have been reported
so far. Recently, it was reported that chrysomelid beetles (Kanarella unicolor Jacobby) feeds
on the leaves of N. nimmoniana without any apparent adverse effect (Ramesha et al., 2011)
(Figure 7c). Interestingly, most of the CPT in the insect body was found in the parental form
without any major metabolized products.

5.1.2 Metabolomics resources used for CPT and structural analogues


For analyzing both the host plants and the isolated endophytes, we employed a number of
state-of-the-art analytical tools and methodologies (Kusari et al., 2009b, 2011b,d). CPT, 9-
MeO-CPT, and 10-OH-CPT were identified by high-performance (pressure) liquid
chromatography (HPLC) coupled to multicomponent high-resolution tandem mass
spectrometry (LC-HRMS and LC-HRMSn fragment spectra) using a LTQ-Orbitrap
spectrometer, Thermo Scientific. The compounds were quantified using TSQ Quantum Ultra
AM mass spectrometer (Thermo Finnigan, U.S.A.) equipped with an ESI ion source (Ion
Max). The mass spectrometer was equipped with a Dionex HPLC system Ultimate 3000
consisting of pump, flow manager, and autosampler (injection volume 0.6 μL). Nitrogen
was used as sheath gas (6 arbitrary units), and helium served as the collision gas. The
separations were performed by using a Phenomenex Gemini C18 column (3 μm, 0.3 × 150
mm) (Torrance, CA) with a H2O (+ 0.1% HCOOH) (A)/acetonitrile (+ 0.1% HCOOH) (B)
gradient (flow rate 4 μL min-1). Samples were analyzed by using a gradient program as
follows: 95% A isocratic for 5 min, linear gradient to 60% A within 12 min, and to 100% B in
29 min. After 100% B isocratic for 5 min, the system returned to its initial condition (95% A)
within 1 min and was equilibrated for 7 min. The spectrometer was operated in positive
252 Metabolomics

mode (1 spectrum s-1; mass range: 200-800) with nominal mass resolving power of 60000 at
m/z 400 with a scan rate of 1 Hz, with automatic gain control to provide high-accuracy mass
measurements within 2 ppm deviation using one internal lock mass; m/z 391.284286; bis-(2-
ethylhexyl)-phthalate. MS2 led to the corresponding CO2 loss of the precursor (CID of 45).
The final MS3 measurement was performed under CID of 45 and resulted in characteristic
fragments of the compounds. The compounds were additionally confirmed using 1H NMR,
performed at 298 K with a Bruker DRX-400 spectrometer using 5 mm tubes with CDCl3
(Merck, Darmstadt, Germany) as solvent.
For the host C. acuminata plants, the LC-MS/MS data were subjected to a number of
different chemometric evaluations for metabolite profiling and correlating the
phytochemical loads among the various Camptotheca plants (infraspecific), among the
organic and aqueous phases, and among the different aerial tissues (dry and fresh in
parallel) to reflect the metabolome profiles of the studied plants (Kusari et al., 2011d). The
analyses included multivariate analysis (MVA), Kruskal’s multidimensional scaling (MDS),
principal component analysis (PCA), linear discriminant analysis (LDA), and hierarchical
agglomerative cluster analysis (HACA). All analyses were performed using the statistical
software XLSTAT-Pro (Addinsoft, NY, U.S.A.), except for MVA which was performed using
the statistical software QI Macros (KnowWare International Inc., CO, U.S.A.). Both the
statistical software packages were used in combination with Microsoft Excel (part of
Microsoft Office Professional, Microsoft Corporation, U.S.A.).
Furthermore, we used the high-precision isotope-ratio mass spectrometry (HP-IRMS) by
compound-specific carbon isotope (CSCI) and compound-specific nitrogen isotope (CSNI)
modules, to confirm that the endophytic fungus actually utilizes host strictosidine
synthase, as detailed above (Kusari et al., 2011b). The CPT produced by the cultured
endophyte (first generation) outside the host plant in a nitrogen-free media was compared
to CPT from the tissue (not containing the same F. solani) of original C. acuminata host
(from SWFU) to check both the δ13C/12C (by CSCI) and the δ15N/14N (by CSNI). It was
possible to trace the exact pattern of the accumulation of both ‘carbons’ and ‘nitrogens’
with the source of the enzyme(s) (fungal or plant) concerned up to and including the
formation of CPT in the endophytic fungus and in the host plant. Briefly, the samples
were readied for HP-IRMS in each case by placing 0.5 mg CPT in 3.5 × 5 mm tin capsules
(HEKAtech GmbH, Germany), lyophilizing completely and finally rolling the capsules
into small spheres. The HP-IRMS measurements were performed in compound-specific
carbon isotope (CSCI) and compound-specific nitrogen isotope (CSNI) modules, using a
FlashEA 1112 elemental analyzer (Thermo Fisher, Italy) coupled to a DELTA V Plus
isotope-ratio mass spectrometer (Thermo Fisher, Bremen, Germany) interfaced through a
ConFlo IV universal continuous flow interface (Thermo Fisher, Bremen, Germany) (Kusari
et al., 2011b). The combustion furnace (oxidation reactor) was maintained at 1020°C, and
flash combustion was initiated by injecting a pulse of O2 at the time of sample drop.
Helium was used as the carrier with a flow rate of 120 mL min-1. NOx species were
reduced to N2 in a reduction furnace at 680°C. Water was removed by phosphorus
pentoxide in a water trap and CO2 was separated from N2 using a Porapak-packed
N2/CO2-separation column (3 m × 6.5 mm, Thermo Electron S. p. A.) operated
isothermally at 85°C. Each sample was analyzed in quadruplet. Acetanilide (Fisons
Instruments) was used as the reference standard.
Metabolomics of Endophytic Fungi Producing
Associated Plant Secondary Metabolites: Progress, Challenges and Opportunities 253

5.1.3 Case study II: Podophyllotoxin and deoxypodophyllotoxin


The first literature report on the extraction of ‘Podophyllum’ was that of King, who called
the resin he obtained from alcohol extraction as ‘podophyllin’ (King, 1857).
‘Podophyllum’ is the dried roots and rhizomes of species of Podophyllum (Figure 8), which
was described and its first modern botanical name given by Linnaeus (1753). The first
successful chemical investigation was later carried out by Podwyssotzki
(1881, 1882, 1884). The correct empirical formula for podophyllotoxin was first advanced
by Borsche and Niemann (1932), and later confirmed (Gensler et al., 1954; Gensler and
Wang, 1954; Petcher et al., 1973).
Strikingly, the first documented proof of the discovery of deoxypodophyllotoxin was not
from Podophyllum. The Leech book of Bald, 900-950 A.D., an early English medicinal book,
has reported on the use of root of Anthriscus sylvestris (Imbert, 1998). These roots were
reported contain lignans such as deoxypodophyllotoxin and were used in ointments
prepared from a large number of plants and plant extracts like savin to cure cancer
(Cockayne, 1961). Podophyllotoxin and deoxypodophyllotoxin share the same action
mechanism based on the core structure of deoxypodophyllotoxin as evidenced by the SAR
studies. They inhibit the formation of the microtubules, i.e., inhibit the formation of the
mitotic spindle, resulting in an arrest of the cell division process in metaphase and clumping
of the chromosomes (Imbert, 1998; Canel et al., 2000; Liu et al., 2007). Under in vitro
conditions, they bind to tubulin dimers giving lignan-tubulin complexes. This stops further
formation of the microtubules at one end but does not stop the disassembly at the other end
leading to the degradation of the microtubules. This mode of action is comparable to the
alkaloid colchicin, and for their mode of action these compounds are called ‘spindle poisons’
(Liu et al., 2007).

Fig. 8. (a) Podophyllum hexandrum Royale. (Photographs courtesy of M. Spiteller). (b)


Podophyllotoxin and some important structural analogues found in plants.
254 Metabolomics

Discoveries of podophyllotoxin-producing endophytic fungi include Phialocephala fortinii


isolated from P. peltatum (Eyberger et al., 2006), Trametes hirsuta isolated from P. hexandrum
(Puri et al., 2006), and Fusarium oxysporum isolated from Juniperus recurva (Kour et al., 2008).
Unfortunately, in all the above cases, there has been no follow-up work on scale-up, and
there is no published breakthrough in the commercial exploitation of these endophytic fungi
as a source of podophyllotoxin. We recently isolated an endophytic fungus, Aspergillus
fumigatus Fresenius, from Juniperus communis sampled from the Rombergpark botanical
gardens, Dortmund, Germany, which produced deoxypodophyllotoxin under in vitro axenic
conditions (Kusari et al., 2009a). The growth and production kinetics showed the potential
of the endophyte in indigenous production of deoxypodophyllotoxin, but in vitro
subculturing showed no production from the third subculture generation.

5.1.4 Metabolomics resources used for podophyllotoxin and structural analogues


The Juniperus and Podophyllum plants sampled by us from diverse populations were
extracted and subjected to metabolomics analyses (Kusari et al., 2011e). HPLC analysis of
the extracts was performed using a Surveyor HPLC system. Compounds were separated on
a Hydro-RP column (150 × 2 mm, 4 μm particle size) from Phenomenex (Torrance, CA). The
mobile phase consisted of 10 mM ammonium acetate in distilled water (A) and acetonitrile
with 0.1% formic acid (B). Gradient elution was performed using the following solvent
gradient: from 85A/15B (held for 3 min) in 16 min to 17A/83B, then in 1 min to 0A/100B
and after 7 min, back to the initial conditions (85A/15B); each run was followed by an
equilibration period of 8 min. The flow rate was 0.22 mL min-1 and the injection volume was
5 μL. All separations were performed at 30°C. Mass spectra were obtained using a TSQ
Quantum Ultra AM mass spectrometer equipped with an ESI ion source (Ion Max)
operating in positive mode (Kusari et al., 2011e). Nitrogen was employed as both the drying
and nebulizer gas (40 AU). Capillary temperature was 200°C and capillary voltage was 3.5
kV. The calibration curves of the available reference compounds podophyllotoxin and
demethylpodophyllotoxin were constructed by dilution of external standards with methanol
to give the desired concentrations. The concentrations of standard solutions were 0.1, 0.5, 1,
5, 10, 50, 80, 120, and 160 μg mL-1. Correlation coefficient for the linear calibration curve was
>0.99 for both podophyllotoxin and demethylpodophyllotoxin. All procedures were carried
out under light protection. Concentrations of the commercially unavailable compounds
deoxypodophyllotoxin and podophyllotoxone were calculated with the assumption of
similar precursor ion response like that of podophyllotoxin. The LOQs were 0.05 μg mL-1
(demethylpodophyllotoxin) and 0.2 μg mL-1 (podophyllotoxin, deoxypodophyllotoxin, and
podophyllotoxone), respectively. The LOD (3 times noise intensities) and LOQ (10 times
noise intensities) were calculated/estimated from signal to noise ratio using signal
intensities of the analytes and the noise near the retention time of the analytes. Estimation
was necessary for the derivatives (deoxypodophyllotoxin and podophyllotoxone) due to
unavailability of reference standards.
All the above secondary metabolites were re-verified using the highly selective and sensitive
LC-ESI-HRMSn. HPLC analysis of the extracts was performed using an Agilent (Santa Clara,
U.S.A.) 1200 HPLC system consisting of LC-pump, PDA detector (λ = 254 nm), autosampler
(injection volume 10 μL) and column oven (30°C). Compounds were separated using a
Synergi Fusion RP80 column (150 x 3 mm, 4 µm particle size) from Phenomenex (Torrance,
Metabolomics of Endophytic Fungi Producing
Associated Plant Secondary Metabolites: Progress, Challenges and Opportunities 255

CA) with a H2O (+ 0.1% HCOOH, + 10 mM ammonium acetate) (A)/acetonitrile (+ 0.1%


HCOOH) (B) gradient (flow rate 400 μL min-1). Samples were analyzed by using gradient
program: 95% A isocratic for 3 min, linear gradient to 100% B over 20 min, after 100% B
isocratic for 10 min, the system returned to its initial condition (95% A) within 1 min, and
was equilibrated for 5 min. The FT-full scan and MS/MS spectra were obtained with an
LTQ-Orbitrap XL spectrometer (Thermo Fisher, U.S.A.) equipped with H-ESI-II source. The
spectrometer was operated in positive mode (1 spectrum s-1; mass range: 250-1000) with
nominal mass resolving power of 60000 at m/z 400 with a scan rate of 1 Hz with automatic
gain control to provide high-accuracy mass measurements within 2 ppm deviation using an
internal standard; bis(2-ethylhexyl)phthalate: m/z 391.284286. MS/MS experiments were
performed in HCD (higher-energy C-trap dissociation, 35 eV) mode. The following
parameters were used for experiments: spray voltage 5 kV, capillary temperature 260°C, and
tube lens 70 V. Nitrogen was used both as sheath gas (45 AU) and auxiliary gas (10 AU).
Helium served as the collision gas.
The LC-MS/MS data were subjected to a number of different chemometric evaluations for
metabolite profiling and correlating the phytochemical loads among the various plants of
the studied Juniperus and Podophyllum species (infraspecific), between the organic and
aqueous extracts, among populations of the same species from different locations, among
populations of different species from the same location, among populations of different
species from different locations, as well as among populations of different genera
(infrageneric) from the same and different locations (Kusari et al., 2011e). The chemometric
algorithms and methodologies were used similar to those used for the C. acuminata plants
(vide supra).
The extracts of the isolated endophytic fungi, both from Juniperus and Podophyllum plants,
were evaluated with an LTQ-Orbitrap spectrometer (Kusari et al., 2009a). The spectrometer
was operated in positive mode (1 spectrum s-1; mass range: 50–1000) with nominal mass
resolving power of 60000 at m/z 400 with a scan rate of 1 Hz with automatic gain control to
provide high accuracy mass within 1 ppm deviation using one internal lock mass,
polydimethylcyclosiloxane – [(CH3)2SiO]6: m/z 445.120025. The spectrometer was equipped
with a Dionex HPLC system Ultimate 3000 consisting of pump, flow manager and
autosampler (injection volume 0.5 μL). Nitrogen was used as sheath gas (5 AU) and helium
served as the collision gas. The separations were performed by using a Phenomenex Gemini
C18 column (3 μm, 0.3 × 150 mm) (Torrance, CA, U.S.A.) with a H2O (+0.1% HCOOH) (A) /
acetonitrile (+0.1% HCOOH) (B) gradient (flow rate 4 μL min-1). Samples were analyzed by
using a gradient program as follows: 90% A isocratic for 2 min, linear gradient to 100% B
over 8 min, after 100% B isocratic for 10 min, the system was returned to its initial condition
(90% A) within 1 min, and was equilibrated for 9 min. Furthermore, for the
deoxypodophyllotoxin-producing endophyte, quantitation of the compound was achieved
by accurate mass (maximum deviation 1 ppm) single ion monitoring (SIM) of the [M+H]+
ion of deoxypodophyllotoxin (Kusari et al., 2009a). Since deoxypodophyllotoxin is
unavailable from commercial sources, the calibration was performed using podophyllotoxin
as standard and detector response was assumed to be in the same range. The calibration
graph was linear from 50 ng mL-1 up to 10000 ng mL-1. Furthermore, a high-resolution full
scan run was performed in order to check for the accumulation of structural analogues of
podophyllotoxin and deoxypodophyllotoxin by that particular endophyte in axenic culture.
256 Metabolomics

5.2 Antidepressants and photodynamic compounds


5.2.1 Case study: Hypericin and emodin
Hypericin (2,2'-dimethyl-4,4',5,5',7,7'-hexahydroxy-mesonaphtodianthrone) (Figure 9), a
naphthodianthrone derivative, is a plant derived compound of high medicinal value. It is one of
the main constituents of Hypericum species. The first detailed report of the isolation of hypericin
was from the medicinal herb Hypericum perforatum L., published by Brockmann et al. (1939). The
molecular formula of hypericin was first reported in 1942 by the same author as C30H16O8
(Brockmann et al., 1942) and eight years later the correct structure was published (Brockmann
et al., 1950). Various species of the genus Hypericum have long been used as medicinal plants in
various parts of the world due to their therapeutic efficacy (Yazaki and Okada, 1994). Their
main constituents are napthodianthrones, primarily represented by hypericin, pseudohypericin,
protohypericin, protopseudophypericin (Brockmann et al., 1939, 1942, 1957), the anthraquinone
emodin, and derivatives occurring in very low concentrations such as isohypericin, demethyl-
pseudohypericin, hyperico-dehydro-dianthrone, pseudo-hyperico-dehydro-dianthrone
(Brockmann et al., 1957), and cyclopseudohypericin (Häberlein et al., 1992). Hypericin has long
been in use, at least from the time of ancient Greece (Tammaro and Xepapadakis, 1986), as an
antidepressant due to its monoamine oxidase (MAO) inhibiting capacity, having effects similar
to bupropion (Nahrstedt and Butterweck, 1997) and imipramine (Raffa, 1998). Potential uses of
hypericin extend to improved wound healing, anti-inflammatory effects (Zaichikova
et al., 1985), antimicrobial and antioxidant activity (Radulovic et al., 2007), sinusitis relief
(Razinkov et al., 1989), and seasonal affective disorder (SAD) relief (Martinez et al., 1993).
Hypericin also has remarkable antiviral activity against a number of viruses (Kusari et al., 2008).
Several recent in vitro studies have revealed the multifaceted cytotoxic activity of hypericin as a
result of its photodynamic activity (Kubin et al., 2005).

Fig. 9. Hypericin and related compounds found in plants.


Metabolomics of Endophytic Fungi Producing
Associated Plant Secondary Metabolites: Progress, Challenges and Opportunities 257

Using the rationale that the plants containing hypericin may also contain endophytic
fungi that are able to accumulate the same or similar molecules, a selective search for
fungal endophytes was pursued. A number of endophytic fungi were isolated from
various organs of the Hypericum plants, which were morphologically different from the
strains isolated from unsterilized explants (surface-contaminating fungi). Only one
endophytic fungus was able to produce hypericin and emodin under axenic submerged
shake-flask fermentation (Kusari et al., 2008). The fungus was identified as Thielavia
subthermophila by its morphology and authenticated by 28S rDNA and ITS-5.8S rDNA
analyses. The growth of the endophyte and production of hypericin remained
independent of the illumination conditions and media spiking with emodin.
Protohypericin could not be detected, irrespective of either spiking or illumination
conditions. The hyp-1 gene, suggested to encode for the Hyp-1 phenolic coupling protein
in plant cell cultures, was absent in the genome of the endophyte. Thus, it was proposed
that emodin anthrone is the common precursor of both hypericin and emodin in the
fungal endophyte, which is governed by a different molecular mechanism than the host
plant or host cell suspension cultures (Kusari et al., 2009c).

5.2.2 Metabolomics resources used for hypericin and related compounds


The Hypericum plants sampled from diverse populations in Europe and the Himalayan
region were extracted and subjected to a metabolic profiling (Kusari et al., 2009d). The
compounds were separated on a Luna C18 100 Å column (3 μm, 250 mm; Phenomenex,
Torrance, CA) at 30°C. Chromatographic conditions were optimized for the separation of
hypericin, pseudohypericin, hyperforin and emodin (gradient 1), and for the separation of
flavonoids (gradient 2) at the Surveyor HPLC system (Thermo Finnigan, U.S.A.). The mobile
phase consisted of 10 mM ammonium acetate buffer adjusted to pH 5.0 with glacial acetic
acid (A) and a 9:1 mixture of acetonitrile and methanol (B). First gradient elution was
performed using the following solvent gradient: from 55A/45B held for 2 min to 0A/100B in
8 min, thereafter holding for 13 min; each run was followed by an equilibration period of 6
min. The flow rate was 0.3 mL min-1 and injection volume was 3 μL. Second gradient elution
was performed using the following solvent gradient: start for 2 min at 95A/5B, in 6 min to
75A/25B, then in 2 min to 50A/50B and in another 2 min to 100B. After holding for 13 min
returned to initial conditions (95A/5B) within 1 min and held for 8 min. The eluent flow rate
was 0.25 mL min-1 and the injection volume was 5 μL. Highly selective and sensitive
selected reaction monitoring (SRM) was performed using a TSQ Quantum Ultra AM mass
spectrometer (Thermo Finnigan, U.S.A.) equipped with an ESI ion source (Ion Max)
operating in negative mode (Kusari et al., 2009d). Nitrogen was employed as both the
drying and nebulizer gas. The capillary voltage was 5 kV and capillary temperature was set
at 200°C. Sheath gas (nitrogen) was set at 45 arbitrary units and collision gas pressure was
1.5 mTorr. Each mass transition was monitored at a peak width of 0.5 and dwell time of 0.3
s. All the secondary metabolites were re-verified using the highly selective and sensitive LC-
ESI-HRMSn (LTQ-Orbitrap spectrometer). External calibration was performed in the range
0.01-10 μg mL-1 for emodin, 0.05-50 μg mL-1 for hyperforin, pseudohypericin, and hypericin,
as well as 0.5-100 μg mL-1 for hyperoside, rutin, quercetin, and quercitrin. Correlation
coefficient for the calibration curves were >0.99 for all analytes. The relative standard
deviation (RSD) of the analytical method was determined by eight injections of an extract
and was below 6% for all compounds. The LOD (limit of detection) and LOQ were
258 Metabolomics

determined by minimum signal to noise ratio of 3 and 9, respectively. Instrumental LOQ of


the compounds varied between 0.003 μg mL-1 (emodin) and 2 μg mL-1 (quercitrin).
Similar to the C. acuminata plants, the LC–MS/MS data for these plants were also subjected
to a number of different statistical evaluations for metabolite profiling and correlating the
phytochemical loads among the various parts of the plants of the studied Hypericum samples
(same and different species), between the organic and aqueous extracts, among the different
species, among populations of the same species from different locations, among populations
of different species from the same locations, as well as among populations of different
species from different locations (Kusari et al., 2009d).
For the isolated endophytic fungi, quantitation of the hypericin and emodin was performed
by using a Thermo Finnigan Surveyor HPLC system consisting of Surveyor MS-pump and
Surveyor Autosampler-Plus (injection volume 5 μL) (Kusari et al., 2008, 2009c). The
compounds were separated on a Luna C18 (50 × 3 mm, 3 μm particle size) column from
Phenomenex (Torrance, CA). The mobile phase consisted of water containing 10 mM
ammonium acetate (pH 5.0) (A) and acetonitrile-methanol, 9:1 (B). Samples were separated
using a gradient program as follows: (flow rate of 250 μL min-1) 55% A isocratic for 2 min,
linear gradient to 100% B over 6 min (flow rate 300 μL min-1). After 100% B isocratic for 7
min, the system was returned to its initial conditions (55% A) within 1 min and was
equilibrated for 4 min before the next run was started. MS detection (multiple reaction
monitoring mode) was performed by using a TSQ Quantum Ultra AM spectrometer
equipped with an ESI ion source (Ion Max) operating in negative mode. Nitrogen was
employed as both the sheath (50 arbitrary units) and auxiliary (8 arbitrary units) gas, and
argon served as the collision gas with a pressure of 1.5 mTorr. The capillary temperature
was set at 250°C. External calibration was performed in the range 0.01-10.0 μg mL-1 for
hypericin and 0.005-10.0 μg mL-1 for emodin. Correlation coefficients for the linear
calibration curves were >0.995 for both compounds.
Hypericin and emodin were identified by HRMS fragment spectra (LTQ-Orbitrap
spectrometer), which were consistent with authentic standards (Kusari et al., 2008, 2009c).
The spectrometer was equipped with a Dionex HPLC system Ultimate 3000 consisting of
pump, flow manager, and autosampler (injection volume 1 μL). Nitrogen was used as
sheath gas (6 arbitrary units), and helium served as the collision gas. The separations were
performed by using a Phenomenex Gemini C18 column (3 μm, 0.3 × 150 mm) (Torrance, CA)
with a H2O (+0.1% HCOOH, +1 mM ammonium acetate) (A)/acetonitrile (+0.1% HCOOH)
(B) gradient (flow rate 4 μL min-1). Samples were analyzed by using a gradient program as
follows: 30% A isocratic for 1 min, linear gradient to 100% B over 10 min; after 100% B
isocratic for 60 min, the system was returned to its initial condition (30% A) within 1 min
and was equilibrated for 9 min. The spectrometer was operated in negative mode (1
spectrum s-1; mass range 50–1000) with nominal mass resolving power of 60000 at m/z 400
with a scan rate of 1 Hz with automatic gain control to provide high-accuracy mass
measurements within 2 ppm deviation using one internal lock mass (m/z 386.7149314; CsI2-).
Additional screening for emodin anthrone and protohypericin was also performed in full
scan negative mode (Kusari et al., 2009c). For that, the spectrometer was equipped with a
Thermo Surveyor system consisting of a LC-pump and autosampler (injection volume 5 μL).
N2 was used as sheath gas (5 arbitrary units), and He served as the collision gas. The
Metabolomics of Endophytic Fungi Producing
Associated Plant Secondary Metabolites: Progress, Challenges and Opportunities 259

separations were performed by using a Phenomenex Synergi Fusion RP column (4 μm, 2 ×


150 mm) with a H2O (+0.1% HCOOH, +10 mM NH4OAc) (A)/MeCN (+0.1% HCOOH) (B)
gradient (flow rate 0.25 mL min-1). Samples were analyzed by using a gradient program as
follows: 50% A isocratic for 2 min, linear gradient to 100% B over 8 min; after 100% B
isocratic for 48 min, the system was returned to its initial condition (50% A) within 1 min
and was equilibrated for 6 min. The spectrometer was operated in negative mode (1
spectrum s-1; mass range 200-1000) with mass resolving power of 60000 at m/z 400 with a
scan rate of 1 Hz with automatic gain control to provide high-accuracy mass measurements
within 2 ppm deviation.

6. Current progress and future challenges


The production of bioactive compounds by endophytes, especially those exclusive to their
host plants, is significant both from the molecular and biochemical perspective, and the
ecological viewpoint. The production of beneficial secondary metabolites (including those
produced by plants) by endophytes nurtures expectations of utilizing them as alternative
and sustainable sources of these compounds. However, the commercial implication of
production of desirable compounds by endophytic fungi still remains a future goal (Kusari
& Spiteller, 2011). A major obstacle preventing the biotechnological application of
endophytes is the perplexing problem of reduction of secondary metabolite production on
repeated subculturing under axenic monoculture conditions. In addition to a constant
pursuit of discovering competent endophytes with potential for pharmaceutical use, it is
essential to follow-up these discoveries with advanced research to establish, restore and
sustain the in vitro biosynthetic capability of endophytes. This can be achieved by a
multifaceted approach involving complete elucidation of the dynamic endophyte-
endophyte interactions pertaining to their biological, biochemical and genetic frameworks.
Considering the fact that endophytes reside within plants and are constantly
communicating with their hosts, it is compelling that plants would have a substantial
influence on the in planta metabolic processes of the endophytes. Moreover, recent whole-
genome sequencing strategies have shown that the known secondary metabolites of various
bacteria and fungi are largely outnumbered by the number of genes encoding the
biosynthetic enzymes in these microorganisms (Winter et al., 2011). This is accentuated by
the fact that endophytic fungi always remain in versatile interactions with the host plant and
other endophytes, and even slight variation in the in vitro cultivation conditions can impact
the kind and range of secondary metabolites they produce (Scherlach & Hertweck, 2009).
Further research to systematically understand the endophyte–endophyte and endophyte-
host interspecies crosstalk is desirable for sustainable production of compounds using
endophytes (Kusari & Spiteller, 2011).

7. Overcoming the obstacles


The potential of novel fungal endophytes capable of biosynthesizing plant metabolites has
undoubtedly been recognized. However, there is still no known breakthrough in the
biotechnological production of these bioactive secondary metabolites using endophytes. It is
important to elucidate the metabolome in endophytes correlating to their associated plants
on a case-by-case basis to understand how the biogenetic gene clusters are regulated and
their expression is affected in planta and ex planta (by environmental changes and axenic
260 Metabolomics

culture conditions). Only a deeper understanding of the host-endophyte relationship at the


molecular and genetic levels might help to induce and optimize secondary metabolite
production under laboratory conditions to yield plant metabolites in a sustained manner
using endophytes. The biosynthesis of plant metabolites in endophytes could further be
manipulated to yield new lead structures which could act as pro-drugs. In addition to
identifying new natural products, genome mining, metabolic engineering and
metagenomics would certainly have an impact on the understanding and manipulation of
secondary metabolite production by endophytic fungi. Likewise, it is well-known that an
unidentified proportion of endophytic fungi are uncultivable in vitro under axenic
conditions. For such unculturable species, environmental PCR strategies might be
employed for recovery, suitably coupled with a culture-independent metagenomic
approach or compound structure-based gene targeting to study their desirable biosynthetic
gene clusters (Kusari & Spiteller, 2011). Further research along these directions is highly
desirable in order to elucidate comprehensively the endophytic fungal biosyntheses since
this knowledge can then be utilized for heterologous expression of the preferred final
products in large quantities using suitable model organisms like Saccharomyces cerevisiae or
Escherichia coli. Another advantage of endophytic fungi over plants is that the biosynthetic
gene modules of a natural product produced by a cascade of biosynthetic steps might be
arranged as an operon in highly contiguous clusters in the fungal genome. This could allow
swift in silico detection of signature genes or gene domains that are pathway-specific
followed by the possibility of expressing them in heterologous organisms. We stand at the
cross-roads of time when the world’s biodiversity is declining at an alarming rate. Many
endemic, endangered and medicinally valuable plants are on the verge of extinction. Along
with these plants, the endophytes harbored in them are also threatened. Further
fundamental research must be addressed to ensure a continuous and sustained supply of
bioactive pro-drugs against the present and emerging diseases.

8. Acknowledgements
We thank the International Bureau (IB) of the German Federal Ministry of Education and
Research (BMBF/DLR), Germany for supporting our various research projects. We also
thank the Ministry of Innovation, Science, Research and Technology of the State of North
Rhine-Westphalia, Germany, and the German Research Foundation (DFG) for granting us
the necessary high-resolution instruments.

9. References
Aly AH, Debbab A, Kjer J, Proksch P. (2010). Fungal endophytes from higher plants: a
prolific source of phytochemicals and other bioactive natural products. Fungal
Divers., 41: 1-16.
Amna T, Puri SC, Verma V, Sharma JP, Khajuria RK, Musarrat J, Spiteller M, Qazi GN.
(2006). Bioreactor studies on the endophytic fungus Entrophospora infrequens for the
production of an anticancer alkaloid camptothecin. Can. J. Microbiol., 52: 189–196.
Anonymous. (2000, 2006). Consideration of proposals for amendment of appendices II: inclusion of
happy tree (Camptotheca acuminata Decaisne) in CITES appendix II of convention in
accordance with the provisions of article II, paragraph 2(a). Prop. 11.58, World
Metabolomics of Endophytic Fungi Producing
Associated Plant Secondary Metabolites: Progress, Challenges and Opportunities 261

Conservation Monitoring Centre, CITES Secretariat/World Conservation


Monitoring Centre, Chatelâine-Genève, Switzerland.
Arnold AE. (2007). Understanding the diversity of foliar endophytic fungi: progress,
challenges, and frontiers. Fungal Biol. Rev., 21: 51-66.
Arnold AE, Henk DA, Eells RA, Lutzoni F, Vilgalys R. (2007). Diversity and phylogenetic
affinities of foliar fungal endophytes in loblolly pine inferred by culturing and
environmental PCR. Mycologia, 99: 185–206.
Arnold AE. (2005). Diversity and ecology of fungal endophytes in tropical forests. In:
Current trends in mycological research, Deshmukh D (ed.), pp. 49-68, Oxford & IBH
Publishing Co. Pvt. Ltd., New Delhi, India.
Arnold AE, Mejia LC, Kyllo D, Rojas EI, Maynard Z, Robbins N, Herre EA. (2003). Fungal
endophytes limit pathogen damage in a tropical tree. Proc. Natl. Acad. Sci. U. S. A.,
100: 15649–15654.
Bacon CW, White JF. (2000). Microbial endophytes, Marcel Deker Inc., New York.
Biswas K, Chattopadhyay I, Banerjee RK, Bandyopadhyay U. (2002). Biological activity and
medicinal properties of neem (Azadirachta indica). Curr. Sci., 82: 1336-1345.
Borsche W, Niemann J. (1932). Über Podophyllin. Justus Liebig’s Ann. Chem., 494: 126–142.
Brockmann H, Falkenhausen EH, Dorlares A. (1950). Die Konstitution des Hypericins.
Naturwissenschaften, 37: 540–540.
Brockmann H, Haschad MN, Maier K, Pohl F. (1939). Über das Hypericin, den
photodynamisch wirksamen Farbstoff aus Hypericum perforatum.
Naturwissenschaften, 27: 550–550.
Brockmann H, Kluge F, Muxfeldt H. (1957). Totalsynthese des Hypericins. Chem. Ber., 90:
2302–2318.
Brockmann H, Pohl F, Maier K, Haschad MN. (1942). Über das Hypericin, den
photodynamischen Farbstoff des Johanniskrautes (Hypericum perforatum). Ann.
Chem., 553: 1–52.
Butterworth JH. (1968). Isolation of a substance that suppresses feeding in locusts. Chem.
Commun., -:23-24.
Canel C, Moraes RM, Dayan FE, Ferreira D. (2000). Podophyllotoxin. Phytochemistry, 54:
115–120.
Cockayne TO. (1961). Leechdoms, wortcunning, and starcraft of early England: being a collection of
documents, for the most part never before printed, illustrating the history of science in this
country before the Norman conquest, vol. 2, The Holland Press, London.
de Bary A. (1866). Morphologie und Physiologie der Pilze, Flechten, und Myxomyceten.
Hofmeister’s handbook of physiological botany, vol. II, Leipzig, Germany.
Decaisne J. (1873). Caracteres et descriptions de trios genres nouveaus de plantes recueilles
en chine par l’abbe a. David Bull. Soc. Bot. France, 20: 155–160.
Eyberger AL, Dondapati R, Porter JR. (2006). Endophyte fungal isolates from Podophyllum
peltatum produce podophyllotoxin. J. Nat. Prod., 69: 1121–1124.
Freeman EM. (1904). The seed-fungus of Lolium temulentum, L., the Darnel. Phil. Trans. R.
Soc. B, 196: 1–27.
Gensler WJ, Wang SY. (1954). Synthesis of picropodophyllin. J. Am. Chem. Soc., 76: 5890–
5891.
Gensler WJ, Samour CM, Wang SY. (1954). Sythesis of a DL-stereoisomer of podophyllic
acid. J. Am. Chem. Soc., 76: 315–316.
262 Metabolomics

Guerin P. (1898). Sur la presence d’un champignon dans l’ivraie. J. Botanique, 12: 230–238.
Gunatilaka AAL. (2006). Natural products from plant-associated microorganisms:
distribution, structural diversity, bioactivity, and implications of their occurrence. J.
Nat. Prod., 69: 509–526.
Gunther RT. (1959). The Greek herbal of Dioscorides. Hafner Publishing Co., New York.
Häberlein H, Tschiersch KP, Stock S, Hölzl J. (1992). Johanniskraut (Hypericum perforatum L.):
Nachweis eines weiteren Naphthodianthrons. Pharm. Ztg. Wiss., 5/137: 169–174.
Hartwell JL, Johnson JM, Fitzgerald DB, Belkin, M. (1953). Podophyllotoxin from Juniperus
species; Savinin. J. Am. Chem. Soc., 75: 235–236.
Hertzberg RP, Busby RW, Caranfa MJ, Holden KG, Johnson RK, Hecht SM, Kingsbury WD.
(1990). Irreversible trapping of the DNA-topoisomerase I covalent complex.
Affinity labeling of the camptothecin binding site. J. Biol. Chem., 265: 19287–19295.
Hickey M, King C. (1981). 100 Families of flowering plants (2nd edition, Walters SM ed.),
Cambridge University Press, Cambridge.
Hölzl J, Petersen M. (2003). Chemical constituents of Hypericum ssp. In: Hypericum: the genus
Hypericum (Series: Medicinal and Aromatic Plants - Industrial Profiles), vol. 31, Ernst E.
(ed.), pp. 77-93, Taylor and Francis, London, UK.
Hombe Gowda HC, Vasudeva R, Mathachen GP, Shaanker RU, Ganeshaiah KN. (2002).
Breeding types in Nothapodytes nimmoniana Graham.: An important medicinal tree.
Curr. Sci., 83: 1077–1078.
Hsiang YH, Hertzberg R, Hecht S, Liu LF. (1985). Camptothecin induces protein-linked
DNA breaks via mammalian DNA topoisomerase I. J. Biol. Chem., 260: 14873–14878.
Imbert TF. (1998). Discovery of podophyllotoxins. Biochimie, 80: 207–222.
Kharwar RN, Verma VC, Kumar A, Gond SK, Harper JK, Hess WM, Lobkovosky E, Ma
C, Ren Y, Strobel GA. (2009). Javanicin, an antibacterial naphthaquinone from an
endophytic fungus of neem, Chloridium sp. Curr. Microbiol., 58: 233-238.
Kharwar RN, Mishra A, Gond SK, Stierle A, Stierle D. (2011). Anticancer compounds
derived from fungal endophytes: their importance and future challenges. Nat. Prod.
Rep., 28: 1208-1228.
King J. (1857). Discovery of podophyllin. Coll. J. M. Sci., 2: 557–559.
Kour A, Shawl AS, Rehman S, Sultan P, Qazi PH, Suden P, Khajuria RK, Verma V. (2008).
Isolation and identification of an endophytic strain of Fusarium oxysporum
producing podophyllotoxin from Juniperus recurva. World J. Microbiol. Biotechnol.,
24: 1115–1121.
Kubin A, Wierrani F, Burner U, Alth G, Grunberger W. (2005). Hypericin - the facts about a
controversial agent. Curr. Pharm. Des., 11: 233–253.
Kumar KR, Ved DK. (2000). 100 Red listed medicinal plants of conservation concern in southern
India, Foundation for Revitalisation of Local Health Traditions (FRLHT), Bangalore,
India.
Kusari S, Kosuth J, Cellarova E, Spiteller M. (2011a). Survival-strategies of endophytic
Fusarium solani against indigenous camptothecin biosynthesis. Fungal Ecol., 4: 219-
223.
Kusari S, Lamshöft M, Spiteller M. (2009a). Aspergillus fumigatus Fresenius, an endophytic
fungus from Juniperus communis L. Horstmann as a novel source of the anticancer
pro-drug deoxypodophyllotoxin. J. Appl. Microbiol., 107: 1019-1030.
Metabolomics of Endophytic Fungi Producing
Associated Plant Secondary Metabolites: Progress, Challenges and Opportunities 263

Kusari S, Lamshöft M, Zühlke S, Spiteller M. (2008). An endophytic fungus from Hypericum


perforatum that produces hypericin. J. Nat. Prod., 71: 159-162.
Kusari S, Spiteller M. (2010). Lessons from endophytes: peering under the skin of plants, In:
Biotechnology – Its Growing Dimensions, Patro, LR (ed.), pp. 1-27, Sonali Publications,
New Delhi, India.
Kusari S, Spiteller M. (2011). Are we ready for industrial production of bioactive plant
secondary metabolites utilizing endophytes? Nat. Prod. Rep., 28: 1203-1207.
Kusari S, Verma VC, Lamshöft M, Spiteller M. (2011c). An endophytic fungus from
Azadirachta indica A. Juss. that produces azadirachtin. World J. Microbiol. Biotechnol.,
in press, doi: 10.1007/s11274-011-0876-2.
Kusari S, Zühlke S, Borsch T, Spiteller M. (2009d). Positive correlations between hypericin
and putative precursors detected in the quantitative secondary metabolite
spectrum of Hypericum. Phytochemistry, 70: 1222-1232.
Kusari S, Zühlke S, Kosuth J, Cellarova E, Spiteller M. (2009c). Light-independent
metabolomics of endophytic Thielavia subthermophila provides insight into microbial
hypericin biosynthesis. J. Nat. Prod., 72: 1825-1835.
Kusari S, Zühlke S, Spiteller M. (2009b). An endophytic fungus from Camptotheca acuminata
that produces camptothecin and analogues. J. Nat. Prod., 72: 2-7.
Kusari S, Zühlke S, Spiteller M. (2011b). Effect of artificial reconstitution of the interaction
between the plant Camptotheca acuminata and the fungal endophyte Fusarium solani
on camptothecin biosynthesis. J. Nat. Prod., 74: 764-775.
Kusari S, Zühlke S, Spiteller M. (2011d). Correlations between camptothecin and related
metabolites in Camptotheca acuminata reveal similar biosynthetic principles and in
planta synergistic effects. Fitoterapia, 82: 497-507.
Kusari S, Zühlke S, Spiteller M. (2011e). Chemometric evaluation of the anti-cancer pro-drug
podophyllotoxin and potential therapeutic analogues in Juniperus and Podophyllum
species. Phytochem. Anal., 22: 128-143.
Lay SV, Denholm AA, Wood A. (1993). The chemistry of azadirachtin. Nat. Prod. Rep., 10:
109-157.
Li GH, Yu ZF, Li X, Wang XB, Zheng LJ, Zhang KQ. (2007). Nematicidal metabolites
produced by the endophytic fungus Geotrichum sp. AL4. Chem. Biodivers., 4: 1520-
1524.
Li S, Zhang Z, Cain A, Wang B, Long M, Taylor J. (2005). Antifungal activity of
camptothecin, trifolin, and hyperoside isolated from Camptotheca acuminata. J. Agric.
Food Chem., 53: 32–37.
Linnaeus C. (1753). Species Plantarum: exhibentes plantas rite cognitas, ad genera relatas, cum
differentiis specificis, nominibus trivialibus, synonymis selectis, locis natalibus, secundum
systema sexuale digestas, vol. 1, Laurentius Salvius, Sweden.
Liu K, Ding X, Deng B, Chen W. (2010). 10-Hydroxycamptothecin produced by a new
endophytic Xylaria sp., M20, from Camptotheca acuminata. Biotechnol. Lett., 32: 689–
693.
Liu YQ, Yang L, Tian X. (2007). Podophyllotoxin: current perspectives. Curr. Bioact. Compd.,
3: 37–66.
Lorence A, Nessler CL. (2004). Camptothecin, over four decades of surprising findings.
Phytochemistry, 65: 2735–2749.
264 Metabolomics

Mahesh B, Tejesvi MV, Nalini MS, Prakash HS, Kini KR, Subbiah V, Hunthrike SS. (2005).
Endophytic mycoflora of inner bark of Azadirachta indica A. Juss. Curr. Sci., 88: 218-
219.
Martinez B, Kasper S, Ruhrmann S, Moller HJ. (1993). Hypericum in the treatment of seasonal
affective disorders. Nervenheilkunde, 36: 103–108.
Nahrstedt A, Butterweck V. (1997). Biologically active and other chemical constituents of the
herb of Hypericum perforatum L. Pharmacopsychiatry, 30: 129–134.
Onelli E, Rivetta A, Giorgi A, Bignami M, Cocucci M, Patrignani G. (2002). Ultrastructural
studies on the developing secretory nodules of Hypericum perforatum. Flora, 197: 92–
102.
Petcher TJ, Weber HP, Kuhn M, von Wartburg A. (1973). Crystal structure and absolute
configuration of 2'-bromopodophyllotoxin-0.5 ethyl acetate. J. Chem. Soc., Perkin
Trans. 2: 288–292.
Podwyssotzki V. (1881). The active constituent of podophyllin. Pharm. J. Trans., 12: 217–218.
Podwyssotzki V. (1882). On the active constituents of podophyllin. Am. J. Pharm., 12: 102–
115.
Podwyssotzki V. (1884). Pharmakologische Studien über Podophyllum peltatum. Naunyn.
Schmied Arch. Exp. Path. Phar., 13: 29–52.
Pommier Y, Kohlhagen G, Kohn KW, Leteurtre F, Wani MC, Wall ME. (1995). Interaction of
an alkylating camptothecin derivative with a DNA base at topoisomerase I-DNA
cleavage sites. Proc. Natl. Acad. Sci. U. S. A., 92: 8861–8865.
Puri SC, Nazir A, Chawla R, Arora R, Riyaz-ul Hasan S, Amna T, Ahmed B, Verma V, Singh
S, Sagar R, Sharma A, Kumar R, Sharma RK, Qazi GN. (2006). The endophytic
fungus Trametes hirsuta as a novel alternative source of podophyllotoxin and
related aryl tetralin lignans. J. Biotechnol., 122: 494–510.
Puri SC, Verma V, Amna T, Qazi GN, Spiteller M. (2005). An endophytic fungus from
Nothapodytes foetida that produces camptothecin. J. Nat. Prod., 68: 1717–1719.
Radulovic N, Stankov-Jovanovic V, Stojanovic G, Smelcerovic A, Spiteller M, Asakawa Y.
(2007). Screening of in vitro antimicrobial and antioxidant activity of nine Hypericum
species from the Balkans. Food Chem., 103: 15–21.
Raffa RB. (1998). Screen of receptor and uptake-site activity of hypericin component of St.
John’s wort reveals sigma receptor binding. Life Sci., 62: 265–270.
Rajagopal R, Suryanarayanan TS. (2000). Isolation of endophytic fungi from leaves of neem
(Azadirachta indica). Curr. Sci., 78: 1375–1378.
Ramesha BT, Zuehlke S, Vijaya R, Priti V, Ravikanth G, Ganeshaiah K, Spiteller M, Shaanker
RU. (2011). Sequestration of camptothecin, an anticancer alkaloid, by chrysomelid
beetles. J. Chem. Ecol., 37: 533-536.
Razinkov SP, Yerofeyeva LN, Khovrina MP, Lazarev AI. (1989). Validation of the use of
Hypericum perforatum medicamentous form with a prolonged action to treat
patients with maxillary sinusitis. Zh. Ushn. Nos. Gorl. Bolezn., 49: 43–46.
Redecker D, Kodner R, Graham LE. Glomalean fungi from the Ordovician. (2000). Science,
289: 1920-1921.
Rehman S, Shawl AS, Kour A, Andrabi R, Sudan P, Sultan P, Verma V, Qazi GN. (2008). An
endophytic Neurospora sp. from Nothapodytes foetida producing camptothecin. Appl.
Biochem. Microbiol., 44: 203–209.
Metabolomics of Endophytic Fungi Producing
Associated Plant Secondary Metabolites: Progress, Challenges and Opportunities 265

Rodriguez RJ, White JFJ, Arnold AE, Redman RS. (2009). Fungal endophytes: diversity and
functional roles. New Phytol., 182: 314–330.
Sankar-Thomas YD. (2010). In vitro culture of Camptotheca acuminata (Decaisne) in Temporary
Immersion System (TIS): growth, development and production of secondary metabolites,
PhD thesis, Universität Hamburg, Germany.
Scherlach K, Hertweck C. (2009). Triggering cryptic natural product biosynthesis in
microorganisms. Org. Biomol. Chem., 7: 1753-1760.
Schulz BJE, Boyle CJC. (2005). The endophytic continuum. Mycol. Res., 109: 661–687.
Sconocchia S. (1983). Scribonius largus compositions. In: Bibliotheca Scriptorum Graecorum et
Romanorum Teubneriana (B.G. Teubner), Hansen GC (ed.), pp. 76-77,
Verlagsgesellschaft, Leipzig, Germany.
Shaanker RU, Ramesha BT, Ravikanth G, Gunaga RP, Vasudeva R, Ganeshaiah, KN. (2008).
Chemical profiling of Nothapodytes nimmoniana for camptothecin, an important
anticancer alkaloid: towards the development of a sustainable production system.
In: Bioactive molecules and medicinal plants, Ramawat KG, Merillon JM (eds.), pp. 197-
213, Springer-Verlag, Berlin and Heidelberg.
Shweta S, Zühlke S, Ramesha BT, Priti V, Kumar PM, Ravikanth G, Spiteller M, Vasudeva R,
Shaanker RU. (2010). Endophytic fungal strains of Fusarium solani, from Apodytes
dimidiata E. Mey. ex Arn (Icacinaceae) produce camptothecin, 10-
hydroxycamptothecin and 9-methoxycamptothecin. Phytochemistry, 71: 117–122.
Staniek A, Woerdenbag HJ, Kayser O. (2008). Endophytes: exploiting biodiversity for the
improvement of natural product-based drug discovery. J. Plant Interact., 3: 75–93.
Stierle A, Strobel GA, Stierle D. (1993). Taxol and taxane production by Taxomyces andreanae,
an endophytic fungus of Pacific yew. Science, 260: 214–216.
Stone JK, Polishook JD, White JF Jr. (2004). Endophytic fungi. In: Biodiversity of fungi:
inventory and monitoring methods, Mueller G, Bills GF, Foster MS (eds.), pp. 241-270,
Elsevier, Burlington, MA, USA.
Strobel GA, Daisy B. (2003). Bioprospecting for microbial endophytes and their natural
products. Microbiol. Mol. Biol. Rev., 67: 491–502.
Strobel GA, Daisy B, Castillo U, Harper J. (2004). Natural products from endophytic
microorganisms. J. Nat. Prod., 67: 257–268.
Sung CK, Kimura T, But PPH, Guo JX. (1998). International collation of traditional and folk
medicine: Northeast Asia, Part III. A project of UNESCO, vol. 3, World Scientific
Publishing Co. Pte. Ltd., Singapore.
Suryanarayanana TS, Thirunavukkarasub N, Govindarajulub MB, Sassec F, Jansend R,
Murali TS. (2009). Fungal endophytes and bioprospecting. Fungal Biol. Rev., 23: 9–
19.
Tammaro F, Xepapadakis G. (1986). Plants used in phytotherapy, cosmetics and dyeing in
the Pramanda district (Epirus, north-west Greece). J. Ethnopharmacol., 16: 167–174.
Veitch GE, Boyer A, Ley SV. (2008). The azadirachtin story. Angew. Chem. Int. Ed., 47: 9402-
9429.
Verma VC, Gond SK, Kumar A, Kharwar RN, Strobel GA. (2007). Endophytic mycoflora of
bark, leaf, and stem tissues of Azadirachta indica A. Juss. (neem) from Varanasi
(India). Microb. Ecol., 54: 119-125.
266 Metabolomics

Verma VC, Gond SK, Mishra A, Kumar A, Kharwar RN, Gange AC. (2009). Endophytic
actinomycetes from Azadirachta indica A. Juss.: isolation, diversity and anti-
microbial activity. Microb. Ecol., 57: 749–756.
Wall ME, Wani MC, Cook CE, Palmer KH, Mcphail AT, Sim GA. (1966). Plant antitumor
agents. I. The isolation and structure of camptothecin, a novel alkaloidal leukemia
and tumor inhibitor from Camptotheca acuminata. J. Am. Chem. Soc., 88: 3888–3890.
Wichtl M. (1986). Hypericum perforatum L. Das Johanniskraut. Zeitschrift Phytother., 3: 87–90.
Winter JM, Behnken S, Hertweck C. (2011). Genomics-inspired discovery of natural
products. Curr. Opin. Chem. Biol., 15: 22-31.
Wu SH, Chen YW, Shao SC, Wang LD, Li ZY, Yang LY, Li SL, Huang R. (2008). Ten-
membered lactones from Phomopsis sp., an endophytic fungus of Azadirachta indica.
J. Nat. Prod., 71: 731–734.
Wu SH, Chen YW, Shao SC, Wang LD, Yu Y, Li ZY, Yang LY, Li SL, Huang R. (2009). Two
new Solanapyrone analogues from the endophytic fungus Nigrospora sp. YB-141 of
Azadirachta indica. Chem. Biodivers., 6: 79-85.
Yazaki K, Okada T. (1994). Hypericum erectum Thunb. (St. John's wort): in vitro culture and
the production of procyanidins. In: Biotechnology in Agriculture and Forestry.
Medicinal and Aromatic Plants VI, Bajaj YPS (ed.), vol. 26, pp. 167-178, Springer-
Verlag, Berlin.
Zaichikova SG, Grinkevich NI, Barabanov EI. (1985). Healing properties and determination
of the upper parameters of toxicity of Hypericum herb. Farmatsiya, 34: 62–64.
Zhang HW, Song YC, Tan RX. (2006). Biology and chemistry of endophytes. Nat. Prod. Rep.,
23: 753–771.
Part 5

Metabolomics in Human Disease Research


11

Metabolomics in the Analysis of


Inflammatory Diseases
Sabrina Kapoor, Martin Fitzpatrick, Elizabeth Clay,
Rachel Bayley, Graham R. Wallace and Stephen P. Young
Rheumatology Research Group, School of Immunity & Infection,
College of Medical and Dental Sciences, University of Birmingham
United Kingdom

1. Introduction
Inflammation is a normal and extraordinarily important component of responses to
infection and injury. The cardinal features of swelling, redness, stiffness and increasing
temperature are strong indicators of the significant changes in tissue metabolism and the
ingress of immune cells into the tissues. The increase in blood flow which underlies
many of these changes may result in changes to the supply of nutrients and in particular
the level of oxygen in the tissues. Inward migration of immune cells, which is also
enabled by the increased blood flow, will put further stress on the metabolic
environment of the tissues. The activity of macrophages and neutrophils in clearing
infection and repairing tissue damage also have significant metabolic consequences
particularly because of the production of cytokines and cytotoxic molecules such as
reactive oxygen species and reactive nitrogen species, which are required to kill invading
organisms. Production of these molecules will consume considerable quantities of
oxygen, ATP and NADPH. These antimicrobial agents put considerable stress on host
cells in the surrounding and distal tissues and can lead to significant loss of protective
metabolites such as glutathione.
Most infections and traumatic injuries are cleared or repaired relatively rapidly and
metabolic homoeostasis is soon restored. However, there is a broad range of inflammatory
diseases which involve chronic activation of the immune system and, as a result, chronic
persistent inflammation. We have been studying the metabolic consequences of chronic
inflammatory diseases with the aim of identifying metabolic fingerprints which may
provide clues about why the localised tissue disease persists. For example, why in
rheumatoid arthritis does persistent inflammation lead to widespread cartilage and joint
destruction? However, the metabolic consequences of chronic inflammation are much
more widespread than the localised disease and can lead on to important comorbidities
such as accelerated atherosclerosis and cardiovascular disease. Metabolomic analysis may
be able to distinguish between localised and systemic metabolic consequences of
inflammation and provide novel targets for therapeutic intervention in these important
human diseases.
270 Metabolomics

2. Introduction to inflammatory disease


An indication of the strong link between inflammation and metabolic processes is seen in
cachexia, the loss of cellular mass associated with disease. The discovery of the involvement
of tumour necrosis factor- alpha (TNF in this process earned it the name ‘cachexin’. While
TNF is now known more generally as a mediator of inflammatory responses, the ability of
inflammatory cytokines to have such profound effects on cellular and metabolic processes is
informative. Systemic inflammation such as that seen in RA causes changes in metabolism
and rheumatoid cachexia is a result of chronic inflammation. This is characterised by the
loss of muscle mass and preservation of fat mass (Evans et al., 2008). Classically cachexia is
characterised by a low BMI. Muscle wasting is a common feature of RA but low BMI is
uncommon as the fat mass is preserved or even increased (Summers et al., 2008). Hence, RA
patients may present with either the classic low BMI cachexia (1-13% of RA population)
(Munro & Capell, 1997) or more frequently, the rheumatoid cachexia (10-20% of RA with
controlled disease and 38% of patients with active RA) (Engvall et al., 2008, Metsios et al.,
2009).
The muscle loss that occurs in rheumatoid cachexia is thought to be due to proinflammatory
cytokines such as TNF, IL1 and IL6. TNF promotes proteolysis through the ubiquitin-
proteasome pathway. There is also some evidence that cytokines may prevent an increase
in muscle protein synthesis in response to feeding (anabolic resistance) (Summers et al.,
2010). In rheumatoid cachexia the degree of muscle wasting is associated with the disease
activity of RA (Summers et al., 2010).

2.1 The inflammatory process


An acute inflammatory reaction is characterised by the classic cardinal signs of
inflammation: heat, redness, swelling and pain. In experimental settings the temporal
relationships oedema, accumulation of leukocytes and accumulation of monocytes and
macrophages are well established. These events in self-limited inflammatory reactions are
coupled with the release of local factors which prevent further release of leukocytes, which
allows resolution (Serhan, 2009). The transition from acute inflammation to chronic
inflammation is widely viewed as a result of an excess of pro-inflammatory mediators.

2.2 Inflammatory mediators


Cytokines are important regulators of inflammation. Some cytokines such as TNF and
interleukin (IL) 1 promote inflammatory responses by inducing cartilage degradation and
promoting a cell-mediated immune response. Other cytokines such as IL-4, IL-10 and IL-13
function mainly as anti-inflammatory molecules (Isomaki & Punnonen, 1997). Key
biological targets that have been identified as being involved in a destructive inflammatory
reaction are COX-2, pro-inflammatory interleukins, TNF, migration inhibition factor,
interferon gamma and matrix metalloproteinases (Ivanenkov et al., 2008).
Several inflammatory mediators have been identified which are common to several
inflammatory diseases. It has been shown that C-reactive protein (CRP) is secreted by several
cell types and is capable of directly activating immune cells. This supports a role for CRP as an
active inflammatory mediator which has systemic and local effects (Montecucco & Mach, 2009).
Metabolomics in the Analysis of Inflammatory Diseases 271

White adipose tissue has been shown to secrete several inflammatory mediators called
adipokines or adipocytokines. These induce their activities by binding to selective
transmembrane receptors. Leptin is the most studied adipocytokine and is thought to have
an important role in the inflammatory process (Montecucco & Mach, 2009).

+ TNF-α
Epithelial
+ IL-17
- IL-10 + IL-1 IL-6
Macrophage IL-1Ra IL-8 IFN-γ
T cell
+ IL-1
- IL-4
IL-6
TNFα + IL-8 IL-10
IL-13
IL-6

Fibroblast
+ FGF
TGF-β

Fig. 1. Key inflammatory cytokines and the inflammatory network. Responses are a balance
of pro-inflammatory tumour necrosis factor alpha (TNF) and interleukin (IL) 1, IL-6, IL-17
and anti-inflammatory IL-1R, IL-4, IL-10 and IL-13. Expression of cytokines is dependent on
activation and local signalling driving progression and eventual resolution.

2.3 Metabolic inflammation


Many factors contribute to the complex course of inflammatory reactions. Microbiological,
immunological and toxic agents can initiate the inflammatory response by activating a variety
of humoral and cellular mediators. In the early phase of inflammation, excessive amounts of
interleukins and lipid-mediators are released and play an important role in the pathogenesis of
organ dysfunction. Arachidonic acid (AA) is released from membrane phospholipids during
inflammatory activation and is metabolised to prostaglandins and leukotrienes. Various
strategies have been evaluated to regulate the excessive production of lipid mediators on
different levels of biochemical pathways, such as inhibition of phospholipase A2, the trigger
enzyme for release of AA, blockade of cyclooxygenase and lipoxygenase pathways and the
development of receptor antagonists against platelet activating factor and leukotrienes. Some
of these agents exert protective effects in different inflammatory disorders such as septic organ
failure, rheumatoid arthritis or asthma, whereas others fail to do so. Encouraging results have
been obtained by dietary supplementation with long chain omega-3 fatty acids like
eicosapentaenoic acid (EPA). In states of inflammation, EPA is released to compete with AA
for enzymatic metabolism inducing the production of less inflammatory and chemotactic
derivatives (Heller et al., 1998).
272 Metabolomics

AA
+ PLA2
EPA -
Prostaglandins
 Leukotrienes

+ COX/LOX Pentaene leukotrienes,


Triene prostaglandins
Proteolysis Thromboxane A3

TNFα +
HIF-1α

+ IL-1 HYPOXIA
IL-6
TNFα
IFNγ
Fig. 2. Some common metabolic responses to inflammation and hypoxia. Arachidonic acid
(AA) from cellular membranes is metabolised to inflammatory prostaglandins and
leukotrienes. Omega-3 fatty acids (EPA) compete for the same pathway producing less
inflammatory derivatives. Hypoxic conditions in the inflammatory site stabilises HIF
transcription factor driving production of IL-1, IL-6, TNFα and IFNγ. TNFα in turn drives
cellular proteolysis and tissue remodelling.

When investigating inflammation it is important to take into account the many facets of the
inflammatory environment that have the potential to play a role in pathology. Hypoxia is
known to be prevalent in the inflammatory environments such as those associated with
wounds, malignant tumours, bacterial infections and autoimmunity (Eltzschig & Carmeliet,
2011, Murdoch et al., 2005). Increasing hypoxia in the inflammatory site is associated with
poorer disease outcome such as increased macroscopic synovitis in rheumatoid arthritis (Ng
et al., 2010).
Normal physiological oxygen levels are thought to range between 5-12% oxygen (compared
to 21% atmospheric oxygen). However, hypoxic tissue oxygen levels in pathological
environments can range from as little as 0.5% oxygen to around 2.5% oxygen. Local hypoxia
develops as the result of either blood vessel occlusion by inflamed tissues, or when existing
supply is insufficient for increased cellular density caused by infiltrating or proliferating
inflammatory cells. Additionally, circulating phagocytes can block blood vessels reducing
blood flow into the inflammatory site (Sitkovsky & Lukashev, 2005). Normal tissue
structures can lend themselves to hypoxia where they are poorly perfused, such as the
synovium or eye. Tissue alteration associated with inflammation can contribute to hypoxia
by altering pressure within the blood vessels causing vessel occlusion and increasing
distances between blood vessels (Jawed et al., 1997, Mapp et al., 1995).
Metabolomics in the Analysis of Inflammatory Diseases 273

There is increasing evidence that the inflammatory environment is hypoxic. The tumour
environment is known to be hypoxic and extensive angiogenesis reveals the requirement of
the tissue for a better oxygen supply. In rheumatoid arthritis, oxygen levels of synovial fluid
have been directly measured revealing lower oxygen tensions compared with osteoarthritic
patients and patients with traumatic joint injuries (Lund-Olesen, 1970). In systemic sclerosis,
direct measurements with sensitive probes revealed lower dermal oxygen levels in fibrotic
areas compared to non-fibrotic areas in both patients and healthy controls (Beyer et al.,
2009). Metabolomic analysis of eye fluids from uveitis patients has shown increased levels
of oxaloacetate and urea, likely derived from anaerobic respiration by locally activated
macrophages (Young et al., 2009, Young & Wallace, 2009).
An elegant cellular oxygen detection system is used by cells to respond to changes in
environmental oxygen. Reductions in environmental oxygen lead to the stabilisation of the
transcription factor hypoxia-inducible factor (HIF), which is otherwise targeted for depletion
in oxygen-rich environments. HIF expression is therefore suggestive of hypoxic exposure, and
has been detected in autoimmune diseases such as rheumatoid arthritis and multiple sclerosis
(Gaber et al., 2009, Hollander et al., 2001, Lassmann, 2003). HIF is known to be important in
inflammatory development, for example loss of HIF-1a in macrophages is associated with
impaired aggregation, motility, invasiveness and killing of bacteria (Cramer et al., 2003).
Hypoxia and HIF stabilisation has a large effect on cellular metabolism. HIF causes a
preference for glycolytic metabolism over oxidative phosphorylation by inducing the
expression of glycolytic enzymes. This allows ATP generation to continue in the absence of
sufficient oxygen albeit at a much reduced efficiency per molecule of glucose. It also induces
the upregulation of lactate dehydrogenase A, therefore promoting the conversion of
pyruvate (produced during glycolysis) to lactate (Wheaton & Chandel, 2011). Lactate has
been detected in many chronic inflammatory conditions such as in inflamed joints (Chang &
Wei, 2011, Treuhaft & McCarty, 1971), multiple sclerosis , pulmonary inflammation (Serkova
et al., 2008) and is thought to play a role in wound healing (Trabold et al., 2003). Conversely,
the acidosis associated with increasing lactate concentrations is thought to play a pathogenic
role in cell transformation and autoantigen development in some inflammatory
environments (Chang & Wei, 2011). Recently, lactate measurements have been suggested to
be useful in the diagnosis of bacterial infections in diabetic foot ulcers compared to non-
infected ulcers. Both infected and non-infected ulcers revealed high lactate concentrations,
but infected ulcers had significantly higher levels probably due to additional immune and
bacterial cell involvement (Loffler et al., 2011). The detection of lactate in metabolomic
studies of disease suggests that there may be an inflammatory component, understanding of
which may help to direct future treatment.
Immune cells are thought to be highly influenced by hypoxia and HIF stabilisation
especially due to the environments they normally act within. In a study performed recently
by Gaber et al., peripheral blood CD4+ T cells placed under hypoxia were found to have a
large induction of genes involved in metabolism and homeostasis (Gaber et al., 2009). Innate
immune cells such as neutrophils and macrophages are thought to be adapted to function
best at lower oxygen tensions as they preferentially use glycolysis to provide ATP even at
higher oxygen levels (Cramer et al., 2003). Macrophages are known to accumulate in the
hypoxic sites of chronic inflammation (Vergadi et al., 2011), and hypoxia is associated with
activation of tissue-resident macrophages. Exposure of macrophages to hypoxic conditions
274 Metabolomics

is associated with upregulation of a whole gamut of proinflammatory cytokines such as IL-1


(Scannell, 1996), IL-6 (Albina et al., 1995), IFN-γ (Murata et al., 2002) and TNF- (White et
al., 2004). It is thought that both low oxygen levels and their downstream effects, such as
lactate production, may give rise to this macrophage phenotype. That such phenotypic
changes are observed in response to the hypoxic conditions of the inflammatory site is
strongly suggestive of a role for metabolism in regulation of immune cells. While normal
wound resolution is a tightly regulated process, the presence of long-term inflammatory
diseases such as rheumatoid arthritis is indicative of the potential for this regulation to go
awry. Therefore hypoxia and the resulting change in metabolism may have a profound
effect on immune cell behaviour and thus influence disease onset and progression.
Adenosine is another molecule produced in response to hypoxia partly by the hypoxic
inhibition of adenosine kinase (Sitkovsky & Lukashev, 2005). It is difficult to detect due to
its local action, but expression of CD39 and CD73, two molecules involved in the
extracellular generation of adenosine, provide a marker of its presence in the inflammatory
environment. Adenosine can have profound effects on immune cells and is generally
perceived to be anti-inflammatory. It is a ligand for specific receptors found on many
immune and stromal cells. These receptors are upregulated by hypoxia suggesting hypoxia
perpetuates both the production and action of this molecule (Hasko et al., 2008, Sitkovsky &
Lukashev, 2005). These receptors have varying downstream effects, with the expression of
the A2A associated with the anti-inflammatory disease but the A2B receptor expression
being implicated in pro-inflammatory conditions such as colitis. Adenosine is known to
cause bronchoconstriction when inhaled by asthma and COPD sufferers, but not in healthy
controls (Hasko et al., 2008). Higher levels of adenosine A2 receptor are seen in asthma
sufferers and these receptors are associated with a pathological role for the molecule in
disease (Brown et al., 2008, Hasko et al., 2008).

2.4 Use of metabolomics in inflammatory diseases


Systemic inflammation causes changes in metabolism and many studies have investigated
individual metabolites in human disease and animal models of inflammation. From these
results it is apparent that the levels of many metabolites are altered by the inflammatory
process and this has provided insights into the mechanisms of disease and uncovered
several potential biomarkers for disease assessment.
Given these profound systemic and localised changes in metabolism provoked by
inflammation and inflammatory cytokines, it is not surprising that metabolomics has been
used to investigate several inflammatory diseases. Metabolomics is able to assess the changes
in several hundred metabolites simultaneously to build disease metabolites profiles. NMR
spectroscopy and mass spectrometry have both been used to derive these multiplexed
metabolite profiles.
These metabolic “fingerprints” have proven useful in discriminating between different
patient groups or identifying responses to therapy, even if the individual metabolites have
not been identified. However, identification of sets of specific metabolites can be derived
from these fingerprints and this has led to the identification of novel biomarkers and novel
pathways in a number of inflammatory diseases. The use of metabolomic analysis of
inflammatory diseases will now be discussed in further detail.
Metabolomics in the Analysis of Inflammatory Diseases 275

3. The inflammatory diseases


3.1 Aetiology
Chronic inflammatory diseases exist in many forms, and have the ability to affect many
systems of the body. These range from localised areas of inflammation such as the gut in
Crohn’s disease, to more widespread systemic inflammation as in rheumatoid arthritis (RA).
Although the mediators and events leading to chronic inflammation are well characterized,
the precise conditions under which acute inflammation becomes chronic are poorly
understood. Recent developments have highlighted the importance of genetic factors,
environmental influences and the interactions between them in the development of chronic
inflammatory disease (Renz et al., 2011).
Research into the genetics of inflammatory disease has been accelerated by genome wide
association studies (GWAS), which has allowed identification of genetic mutations
associated with an increased risk of developing specific conditions. For example, many
immunologically relevant genes have been associated with an increased risk of developing
RA. These include human leukocyte antigen (HLA) alleles involved in antigen recognition,
and the peptidyl-arginine deiminase type IV (PADI4) gene controlling production of cyclic
citrullinated proteins (CCP’s) commonly seen in RA (Nishimoto et al., 2010). However, in a
complex disease like RA, genetics are not the whole story, as illustrated by the fact that twin
studies only report a concordance rate of around 60% (MacGregor et al., 2000). Thus the
importance of external environmental factors in the development of inflammatory diseases
should be considered.
Chronic inflammatory diseases have become more prevalent in recent years, and as major
genetic changes are unlikely to have occurred over such a short time period, this is likely to
be a result of alterations in environmental exposures and lifestyle factors. To date, several
factors have been identified as significant contributors including ageing, infection, poor
nutrition and smoking.
Smoking raises an individual’s risk of developing inflammatory disease considerably. It has
numerous effects on the body including activation of the acute inflammatory response and
introduction of large amounts of reactive oxygen species (ROS) (Borgerding & Klus, 2005). It
is unclear as to which particular constituent of smoke induces the inflammatory response;
however studies have revealed that smoke contains large amounts of lipopolysaccharide
(LPS) (Hasday et al., 1999), which could potentially trigger unwanted immune responses
seen in chronic inflammatory disease. An increase in ROS is also evident, as indicated by
decreased circulating antioxidants found in smokers (Alberg, 2002). This creates a pro-
oxidant environment and increases the likelihood of oxidative damage to important cellular
components.
It is not surprising given the complex and varied nature of chronic inflammatory diseases
that the observed phenotype is a result of gene-gene and gene-environment interactions. For
example, it has been shown in mice with a mutation in the Crohn's disease (CD)
susceptibility gene Atg16L1 who become infected with murine norovirus develop a Crohn’s-
like disease (Stappenbeck et al., 2010). There was no evidence of pathology in the wild type
mouse, suggesting the presence of two risk factors is required to induce disease. Another
example of gene-environment interactions in disease development was found when looking
276 Metabolomics

at the interaction between RA susceptibility genes HLA-DRB1 and PTPN22 and their
interaction with smoking (Kallberg et al., 2007). It was observed that the odds ratio (OR) of
developing RA with two genetic risk factors was 13.2, which rose to 23.4 if two genetic
factors were present and there was a history of smoking. These studies provides sound
evidence that gene-gene and gene-environment interactions occur, and risk of inflammatory
disease greatly increases with the presence of more than one additional risk factor.

3.2 The gut


Crohn’s disease is a chronic debilitating inflammatory disease of the bowel. The exact
aetiology is unknown but is thought to be related to the dysregulation of the immune
response towards gut microflora (Strober et al., 2007). Urinary metabolite profiling was
carried out on a mouse model of Crohn’s disease. These samples were analysed using gas
chromatography-mass spectrometry and five key metabolic differences were identified
between the Crohn’s disease model and controls. This suggested that there are alterations of
tryptophan metabolism, fucosylation and fatty acid metabolism in Crohn’s disease mice and
the authors concluded that fucose and xanthurenic acid could be useful markers of gut
inflammation (Lin et al., 2009).
Using a mouse model of inflammatory bowel disease (IBD) to investigate urinary
metabolites using NMR, it was found that there was an increase in trimethylamine (TMA)
and fucose compared to controls. The increase in TMA was parallel to the progression of
IBD (Murdoch et al., 2008). A mouse model of Ulcerative Colitis (UC) was used to looked at
serum and urinary metabolites (Schicho et al., 2010). These authors found that both serum
and urine were equally powerful for detecting colitis but the metabolites responsible for the
differences were different for serum and urine.
Metabolomics of faecal extracts have also been used to study inflammatory bowel disease
(Bezabeh et al., 2009). It is sometimes difficult to distinguish Crohn’s disease (CD) from UC
and some cases are labelled as indeterminate. Over time these cases are usually identified
by a combination of endoscopic, radiological and histological techniques. Earlier
identification could aid treatment and prognostication. Metabolomic analysis of faecal
extracts of patients with both inflammatory diseases showed reduced levels of butyrate,
acetate, methylamine and TMA compared to control (Marchesi et al., 2007). Comparing the
UC and CD samples glycerol, alanine, isoleucine, leucine, lysine and valine were present in
higher quantities in CD compared to UC. Acetate was lower in CD compared to UC
(Marchesi et al., 2007). Metabolic differences were more marked in CD indicating that
inflammation is more extensive in CD compared to UC.
Urinary metabolites have also been used to distinguish CD and UC in humans (Williams et
al., 2009). They found that specific urinary metabolites related to gut metabolism differed
between CD, UC and controls. Hippurate was lowest in CD and differed significantly
between CD, UC and controls. Formate levels were higher in CD than in UC or controls and
4-cresol sulphate was lower in CD than in UC or controls (Williams et al., 2009). Hippurate
has been shown to be modulated according to gut microbes and this difference is likely to
reflect changes in intestinal microbes.
In summary several studies have looked at IBD. The studies have shown that both in mice
and in humans TMA is an important marker of IBD (Marchesi et al., 2007, Murdoch et al.,
Metabolomics in the Analysis of Inflammatory Diseases 277

2008, Schicho et al., 2010). This has been shown using both urine samples or faecal extracts.
Hence, TMA may be a useful biomarker for IBD.

3.3 The eye


As a closed and immuno-privileged site, the eye provides an ideal system for metabolic
analysis. Metabolic products of inflammatory infiltrate accumulate in the vitreous fluid of
the eye and may be extracted during other corrective surgery.
Metabolomics has been used to look at vitreous humour in order to differentiate ocular
inflammatory diseases (Young et al., 2009). Vitreous fluid samples were taken from patients
undergoing retinal surgery and analysed using NMR. Patients had various retinal disorders
including chronic non-infectious uveitis (CU), lens-induced uveitis (LIU), proliferative diabetic
retinopathy, proliferative vitreoretinopathy (PVR), rhegmatogenous retinal detachment,
candida endopthalmitis and varicella zoster virus acute retinal necrosis. The different disease
groups showed clear separation using principle component analysis (PCA) and partial least
squared discriminate analysis (PLSDA). The majority of the patients had LIU and CU. When
looking at LIU and CU specifically there was clear separation and individual metabolites from
the spectra showed significant differences with urea, oxaloacetate and glucose all being raised
in LIU compared to CU. As urea and oxaloacetate are both involved in the urea cycle it
suggests that there is more active inflammation in the LIU patients (Young et al., 2009).
NMR has also been used to look at ocular metabolism in pig eyes (Greiner et al., 1985). They
used phosphorous NMR and found phosphorous containing metabolites in aqueous and
vitreous fluids (Greiner et al., 1985). In addition to quantifying metabolites, phosphorous
NMR can be used to monitor the rate of metabolic change in a specific biochemical reaction
and the rate of change in the concentration of a particular metabolite (Greiner et al., 1985).
Phosphorous NMR provides a non-invasive method to analyse ocular tissues metabolically
and detect subtle biochemical changes that precede manifestations of disease. Such
detection may allow for early and more effective therapeutic intervention.

3.4 Neurological disease


Multiple sclerosis (MS) is a chronic inflammatory disease affecting the nervous system. Its
aetiology is still not completely understood (Ibrahim & Gold, 2005). It is characterised by
demyelination, axonal loss and breakdown of the blood-brain barrier (Trapp et al., 1999). It
is a heterogeneous, relapsing and remitting disease. Different treatments have been shown
to work at different stages of disease (Rieckmann & Smith, 2001) so it is important to
identify biomarkers that enable identification of different phases.
Interleukin-1β (IL-1β) and TNF-α, have been found to be associated with a broad spectrum
of neurological diseases including MS. Griffin et al looked at rat urines to determine
whether NMR spectroscopy could detect the presence of IL-1β and TNF-α induced lesions
and distinguish between the pathology caused (Griffin et al., 2004). They used an
adenoviral vector to induce chronic endogenous expression of either IL-1β or TNF-α. They
found significant differences between the groups, with the IL-1β treated group showing
increases in leucine, isoleucine, valine, n-butyrate and glucose whilst the TNF-α treated
group showed increases of citrate, 2-oxoglutarate and succinate (Griffin et al., 2004).
278 Metabolomics

NMR spectroscopy has also been used to assess cerebrospinal fluid (CSF) in patients with
MS. It has been shown that there are increased CSF levels of lactate, creatinine and
fructose in MS compared to control patients (Nicoli et al., 1996). Two additional
unidentified signals were found to be elevated in MS. The compound responsible for
both these signals has now been identified as B-hydroxyisobutyrate (Lutz et al., 2007).
This is a typical partial degradation product of branched-chain amino acids. Increased B-
hydroxyisobutyrate in urine is thought to be due to respiratory-chain deficiency leading
to impaired oxidation of NADH (Chitayat et al., 1992). However the level of B-
hydroxyisobutyrate in these experiments was much higher than the level found in CSF
from MS patients, and so the precise role of B-hydroxyisobutyrate in MS needs further
investigation.
In a study of metabolite fingerprints in the CSF from patients with a range of neurological
conditions we have been able to differentiate between some of these conditions by
comparing the metabolites found (Sinclair et al., 2010). In particular we were able to identify
some novel features of idiopathic intracranial hypertension (IIH) a neurological condition,
the pathogenesis of which is poorly understood (Sinclair et al., 2008). Although IIH was not
thought to be an inflammatory disease, the elevated levels of lactate we observed in IIH
points towards an inflammatory component since lactate has been identified in
inflammatory CNS disease previously (Simone et al., 1996). Rabbits with elevated
intraocular pressure also show increased levels of lactate which may reflect anaerobic
metabolism resulting from decreased blood supply and this may also be an explanation for
the lactate in the IIH patients’ CSF due to compressed vasculature from the elevated
intracranial pressure. Oxaloacetate levels were also increased in IIH and this, together with
reduced citrate, suggests alterations in the citric acid cycle. Overall the observations suggest
a predominantly anaerobic environment deficient in carbohydrate substrate in patients with
IIH, a conclusion supported by the presence of elevated ketone bodies 3-hydroxybutyrate
(Sinclair et al., 2010) often observed in hypoxic tissues.

3.5 Lung disease


Pulmonary inflammation contributes to the pathogenesis of a number of lung diseases.
There is a growing need for validated experimental models that can help our understanding
of disease pathogenesis and therapeutic intervention. Traditionally animal models have
been used but they have their own problems in representing human disease. Genetic
manipulation can greatly enhance animal models. NMR has had some application in the
quantification of experimental lung injury.
Serkova et al used Magnetic Resonance Imaging (MRI) and NMR to try and detect and
quantify injury in mice following intratracheal administration of inflammatory cytokines
(Serkova et al., 2008). Pulmonary inflammation was induced by intratracheal administration
of IL-1β and TNF-α. Lung tissue was used for the NMR metabolomics. They showed that
with pulmonary inflammation there was a 50% depletion of ATP and a corresponding
elevation of the lactate to glucose ratio suggesting a shift to anaerobic metabolism during
inflammation. These returned to control levels at 24 hours (Serkova et al., 2008). These data
show that intratracheal administration of IL-1β and TNF-α leads to profound but reversible
pulmonary inflammation which is detectable by NMR.
Metabolomics in the Analysis of Inflammatory Diseases 279

3.6 Osteoarthritis
Osteoarthritis (OA) is a complex disease and has a multifactorial pathogenesis. It has many
known risk factors such as age, sex, obesity, activity level, prior joint damage and genetic
susceptibility. It is not classically thought of as an inflammatory disease but it may have an
inflammatory element. There are currently no disease-modifying drugs for OA and very
few are in development.
Synovial fluid (SF) has been used to look at OA via NMR. SF is felt to be a good medium to
study as the SF is the first place where the degradation products, enzymes and signal
transduction molecules involved in OA are released from the cartilage matrix. The SF
should therefore have a higher concentration of metabolites compared to blood, lymph or
urine.
Damayanovich et al used SF from a canine model of OA to look at metabolic profiles
using NMR (Damyanovich et al., 1999). Metabolites from experimentally induced canine
knee OA SF were compared to metabolites from SF of normal canine knees. They found
large increases in lactate and sharp decreases of glucose in OA SF compared to normal
SF suggesting that the intra-articular environment of an OA joint is more hypoxic and
acidic than a healthy joint. They also found increased levels of pyruvate, lipoprotein
associated fatty acids, glycerol and ketones in OA SF suggesting that lipolysis may be an
important source of energy in OA. There were also elevated levels of N-
acetylglycoproteins, acetate and acetamide in OA SF especially with progressive OA
(Damyanovich et al., 1999).
In order to understand further the mechanisms behind OA progression, Damayanovich et al
looked at the effect of joint afferent nerve injury (Damyanovich et al., 1999). They again
used a bilateral canine model of OA. Paired SF samples were taken from dogs that had
undergone bilateral anterior cruciate ligament transaction, unilateral knee denervation and
contralateral sham nerve exposure. NMR was used to look at the SF. Increases in glycerol,
hydroxybutyrate, glutamine, creatinine, acetate and N-acetyl-glycoprotein were seen in the
SF from denervated compared to control knees. This suggests that the metabolite
differences seen in the denervated knees are due to the aggravation of OA caused by joint
denervation (Damyanovich et al., 1999). Hydroxybutyrate is also found in SF of RA patients
(Naughton et al., 1993) suggesting that it is more of a marker of joint destruction rather than
being specific for any joint disease.
Another group used guinea pigs to study OA metabolism (Lamers et al., 2003). They used
Hartley outbred strain guinea pigs as they develop spontaneous progressive knee OA
with features similar to human disease. The earliest histological features appear at 3
months but progress to extensive cartilage degeneration after 12 months. Urine samples
were collected from these OA guinea pigs and from healthy animals at 10 and 12 months
of age. They identified a metabolic fingerprint that reflected OA changes in the pigs.
Lactic acid, malic acid, hypoxanthine and alanine contributed strongly to the fingerprint
suggesting their involvement in OA (Lamers et al., 2003). The metabolic profile largely
resembled that found in the guinea pig model. The presence of hypoxanthine suggests
that OA may be an inflammatory disease due to the increased oxygen demand and altered
purine metabolism.
280 Metabolomics

Mass spectroscopy has also been used to look for novel biomarkers for knee OA (Zhai et al.,
2010). They looked at serum samples of unrelated white women with and without knee OA.
Knee OA was defined as radiographic, medically diagnosed or total knee replacement due
to primary OA. They found that the ratio of valine to histidine and the ratio of leucine to
histidine to be significantly associated with knee OA in humans (Zhai et al., 2010). These
ratios have potential clinical use as an OA biomarker. OA branched chain amino acids
(BCAA) are raised which may drive the release of acetoacetate and 3-hydroxybutyrate.
These can result from the partial oxidation of leucine. BCAA are essential amino acids and
therefore cannot be synthesised within the body. An increased level of BCAA may suggest
an increased rate of protein breakdown or be secondary to collagen degradation. BCAA
increase production of the cytokines IL1, IL2, TNF and interferon (Bassit et al., 2000) which
could drive the collagen degradation.

3.7 Rheumatoid arthritis


Rheumatoid arthritis (RA) is a debilitating systemic inflammatory joint disease. An
abnormal metabolic profile in the inflamed joint in RA may be due to the impairment of the
vascular supply and/or an increase in the metabolic rate of the inflamed joint.
Hyaluronic acid is a major component of the proteoglycan aggregate of articular cartilage
which is required for the functional integrity of extracellular matrix. In RA, SF hyaluronate
is depolymerised by the action of reactive oxygen radical species (Parkes et al., 1991).
Hyaluronidase activity is absent in both normal and inflamed SF. Generation of reactive
oxygen species plays a principal part in synovial hypoxic reperfusion injury (Farrell et al.,
1992). This occurs as increased intra-articular pressure during exercise exceeds synovial
capillary perfusion pressure leading to impaired blood flow (Mapp et al., 1995).
In 1993, The Inflammation Research Group, The London Hospital Medical College looked at
the NMR profiles of RA SF and matched serum samples (Naughton et al., 1993). The NMR
profiles of SF were markedly different from their matched serum samples. There were high
levels of lactate in the SF compared to the serum and low levels of glucose in the SF
compared to the serum. These changes are consistent with the hypoxic status of the
rheumatoid joint (Naughton et al., 1993). All the SF samples (RA and control) had lower
levels of chylomicron and very-low-density-lipoprotein associated triglycerides compared
to their matched serum samples. The SF samples also had high levels of ketone bodies
compared to their matched serum samples. These results suggest that the intra-articular
environment has an increased utilisation of fats for energy even though it is hypoxic
(Naughton et al., 1993, Naughton et al., 1993). They were unable to compare the control SF
to the rheumatoid SF due to the low levels of SF aspirated.
Serum from mice has been used to identify a metabolite biomarker pattern associated with
RA (Weljie et al., 2007). Using NMR they found that uracil, xanthine and glycine could be
used to distinguish arthritic from control animals (Weljie et al., 2007). The presence of the
metabolites suggests that nucleic acid metabolism may be highly affected in RA and there
may be an association with oxidative stress.
More recently, a group in Denmark have looked at the plasma of patients with RA (Lauridsen
et al., 2010). They found differences in the metabolites between patients with RA and healthy
controls and differences between patients with active RA and controlled RA. The metabolites
Metabolomics in the Analysis of Inflammatory Diseases 281

that they identified were cholesterol, lactate, acetylated glycoprotein and lipids. The lactate
levels represented oxidative damage and thus indirectly reflected active inflammation.

3.8 Atherosclerosis
Atherosclerosis is the thickening of arteries and is the underlying pathological process that
affects the coronary, cerebral, aortic and peripheral arteries. Atherosclerosis involves the
accumulation of cholesterol particles, cellular by-products, deposition of the extracellular
matrix and inflammatory cell infiltration within the vessel wall (Goonewardena et al., 2010).
Chronic inflammation has been recognised as one of the key components of atherogenesis
(Ross, 1999) but accelerated atherosclerosis is an important confounder of chronic
inflammatory diseases such as rheumatoid arthritis (Bacon et al., 2005). Animal models have
been widely used to investigate the biochemical basis of atherosclerosis. Using aortas from
apolipoprotein-E knockout mice Mayr et al concluded that inefficient vascular glucose and
energy metabolism coincided with increased oxidative stress in animals with hyperlipidaemia
(Mayr et al., 2007). NMR-based metabolomics of mouse urine has been used to look at
atherosclerosis (Leo & Darrow, 2009). Using apolipoprotein-E knockout mice they compared
untreated mice with those treated with captopril. They found elevated levels of xanthine and
ascorbate in untreated mice which may be possible markers of plaque formation (Leo &
Darrow, 2009). The interaction between diet and inflammation in promoting atherosclerosis
has also been highlighted through metabolomic studies and Kleenmann (Kleemann et al.,
2007) suggested that a high cholesterol intake lead to a switch in liver metabolism towards a
pro-atherosclerotic state. Another recent example of how metabolomics can provide novel
insights into inflammatory disease pathology was the observation that the metabolism of
dietary lecithin by gut flora leads to the increased absorption and accumulation of choline
derivatives which in turn promote cardiovascular disease (Wang et al., 2011) . Only through
the use of the systematic analysis of metabolites using metabolomics was it possible to uncover
these complex metabolic relationships underpinning the disease process.

4. Conclusion
As summarised above there is now a growing body of literature describing metabolomic
changes in inflammatory diseases, both in humans and animal models. Several distinct
metabolic changes have been identified in inflammatory disorders, but there is a core theme
of increasing energy requirements coupled with decreasing oxygen supply within the
inflammatory environment.
Studies in MS, RA, OA and inflammatory lung disease have all shown an increase in lactate,
while studies of inflammatory eye and lung diseases have shown local reductions in
glucose. Immunological responses to tissue hypoxia, such as the up-regulation of IL-1, IL-6,
IFN-γ and TNF-α seen in macrophages, show the link between local metabolic changes and
inflammatory responses. Here transcription factor HIF-1α may play a central co-ordinating
role in both normal and pathological inflammation by regulating the underlying cellular
metabolism towards anaerobic respiratory pathways and lactate production. Subsequent
effects of inflammatory cytokines on tissue remodelling and perfusion further provide a
mechanism for feedback driving self-sustaining inflammatory microenvironments, and
potentially where resolution is disrupted, a route to chronic inflammatory disease.
282 Metabolomics

Therefore, as both a by-product and mediator of local tissue conditions, metabolites offer a
unique opportunity to gain an insight of local and global inflammatory processes.
Metabolomics likewise, provides promising opportunities for both diagnosis of
inflammatory diseases, and study of the underlying processes that may offer clues as to how
the inflammatory process develops.

5. Acknowledgement
The authors were supported by grants from Arthritis Research UK (grant numbers 18552
and 19325) and the Wellcome Trust (089384/Z/09/Z and 066490/Z/01/A).

6. References
Alberg, A.J. (2002). The influence of cigarette smoking on circulating concentrations of
antioxidant micronutrients, Toxicology, vol.180, No. 2, pp.121-37, ISSN 0300-483X.
Albina, J.E., Henry, W.L., Jr., Mastrofrancesco, B., Martin, B.A., Reichner, J.S. (1995).
Macrophage activation by culture in an anoxic environment, Journal of Immunology,
vol.155, No. 9, pp.4391-6, ISSN 0022-1767.
Bacon, P.A., Church, L.D., Young, S.P. (2005). Endothelial Dysfunction - the Link Between
Inflammation and Atherosclerosis in Rheumatoid Arthritis, Journal of the Indian
Rheumatology Association, vol.13, pp.103-6, ISSN 0971-5045.
Bassit, R.A., Sawada, L.A., Bacurau, R.F.P., Navarro, F., Rosa, L.F.B.P. (2000). The effect of
BCAA supplementation upon the immune response of triathletes, Medicine and
Science in Sports and Exercise, vol.32, No. 7, pp.1214-9, ISSN 0195-9131.
Beyer, C., Schett, G., Gay, S., Distler, O., Distler, J.H. (2009). Hypoxia. Hypoxia in the
pathogenesis of systemic sclerosis, Arthritis Research and Therapy, vol.11, No. 2,
pp.220, ISSN 1478-6362.
Bezabeh, T., Somorjai, R.L., Smith, I.C.P. (2009). MR metabolomics of fecal extracts:
applications in the study of bowel diseases, Magnetic Resonance in Chemistry, vol.47,
pp.S54-S61, ISSN 0749-1581.
Borgerding, M., Klus, H. (2005). Analysis of complex mixtures - Cigarette smoke,
Experimental and Toxicologic Pathology, vol.57, pp.43-73, ISSN 0940-2993.
Brown, R.A., Spina, D., Page, C.P. (2008). Adenosine receptors and asthma, British Journal of
Pharmacology, vol.153 Suppl 1, pp.S446-S56, ISSN 0007-1188.
Chang, X., Wei, C. (2011). Glycolysis and rheumatoid arthritis, International Journal of
Rheumatic Diseases, vol.14, No. 3, pp.217-22, ISSN 1756-185X.
Chitayat, D., Meaghervillemure, K., Mamer, O.A., Ogorman, A., Hoar, D.I., Silver, K.,
Scriver, C.R. (1992). Brain Dysgenesis and Congenital Intracerebral Calcification
Associated with 3-Hydroxyisobutyric Aciduria, Journal of Pediatrics, vol.121, No. 1,
pp.86-9, ISSN 0022-3476.
Cramer, T., Yamanishi, Y., Clausen, B.E., Forster, I., Pawlinski, R., Mackman, N., et al. (2003).
HIF-1alpha is essential for myeloid cell-mediated inflammation, Cell, vol.112, No.
5, pp.645-57, ISSN 0092-8674.
Damyanovich, A.Z., Staples, J.R., Chan, A.D.M., Marshall, K.W. (1999). Comparative study
of normal and osteoarthritic canine synovial fluid using 500 MHz H-1 magnetic
Metabolomics in the Analysis of Inflammatory Diseases 283

resonance spectroscopy, Journal of Orthopaedic Research, vol.17, No. 2, pp.223-31,


ISSN 0736-0266.
Damyanovich, A.Z., Staples, J.R., Marshall, K.W. (1999). H-1 NMR investigation of changes
in the metabolic profile of synovial fluid in bilateral canine osteoarthritis with
unilateral joint denervation, Osteoarthritis and Cartilage, vol.7, No. 2, pp.165-72,
ISSN 1063-4584.
Eltzschig, H.K., Carmeliet, P. (2011). Hypoxia and Inflammation REPLY, New England
Journal of Medicine, vol.364, No. 20, pp.1977-, ISSN 0028-4793.
Engvall, I.L., Elkan, A.C., Tengstrand, B., Cederholm, T., Brismar, K., Hafstrom, I. (2008).
Cachexia in rheumatoid arthritis is associated with inflammatory activity, physical
disability, and low bioavailable insulin-like growth factor, Scandinavian Journal of
Rheumatology, vol.37, No. 5, pp.321-8, ISSN 0300-9742.
Evans, W.J., Morley, J.E., Argiles, J., Bales, C., Baracos, V., Guttridge, D., et al. (2008).
Cachexia: A new definition, Clinical Nutrition, vol.27, No. 6, pp.793-9, ISSN 0261-
5614.
Farrell, A.J., Williams, R.B., Stevens, C.R., Lawrie, A.S., Cox, N.L., Blake, D.R. (1992).
Exercise Induced Release of Vonwillebrand-Factor - Evidence for Hypoxic
Reperfusion Microvascular Injury in Rheumatoid-Arthritis, Annals of the Rheumatic
Diseases, vol.51, No. 10, pp.1117-22, ISSN 0003-4967.
Gaber, T., Haupl, T., Sandig, G., Tykwinska, K., Fangradt, M., Tschirschmann, M., et al.
(2009). Adaptation of human CD4+ T cells to pathophysiological hypoxia: a
transcriptome analysis, Journal of Rheumatology, vol.36, No. 12, pp.2655-69, ISSN
0315-162X.
Goonewardena, S.N., Prevette, L.E., Desai, A.A. (2010). Metabolomics and atherosclerosis,
Current Atherosclerosis Reports, vol.12, No. 4, pp.267-72, ISSN 1534-6242.
Greiner, J.V., Kopp, S.J., Glonek, T. (1985). Phosphorus Nuclear-Magnetic-Resonance and
Ocular Metabolism, Survey of Ophthalmology, vol.30, No. 3, pp.189-202, ISSN 0039-
6257.
Griffin, J.L., Anthony, D.C., Campbell, S.J., Gauldie, J., Pitossi, F., Styles, P., Sibson, N.R.
(2004). Study of cytokine induced neuropathology by high resolution proton NMR
spectroscopy of rat urine, FEBS Letters, vol.568, No. 1-3, pp.49-54, ISSN 0014-5793.
Hasday, J.D., Bascom, R., Costa, J.J., Fitzgerald, T., Dubin, W. (1999). Bacterial endotoxin is
an active component of cigarette smoke, Chest, vol.115, No. 3, pp.829-35, ISSN
0012-3692.
Hasko, G., Linden, J., Cronstein, B., Pacher, P. (2008). Adenosine receptors: therapeutic
aspects for inflammatory and immune diseases, Nature Reviews Drug Discovery,
vol.7, No. 9, pp.759-70, ISSN 1474-1784.
Heller, A., Koch, T., Schmeck, J., van Ackern, K. (1998). Lipid mediators in inflammatory
disorders, Drugs, vol.55, No. 4, pp.487-96, ISSN 0012-6667.
Hollander, A.P., Corke, K.P., Freemont, A.J., Lewis, C.E. (2001). Expression of hypoxia-
inducible factor 1alpha by macrophages in the rheumatoid synovium: implications
for targeting of therapeutic genes to the inflamed joint, Arthritis and Rheumatism,
vol.44, No. 7, pp.1540-4, ISSN 0004-3591.
284 Metabolomics

Ibrahim, S.M., Gold, R. (2005). Genomics, proteomics, metabolomics: what is in a word for
multiple sclerosis?, Current Opinion in Neurology, vol.18, No. 3, pp.231-5, ISSN
1350-7540.
Isomaki, P., Punnonen, J. (1997). Pro- and anti-inflammatory cytokines in rheumatoid
arthritis, Annals of Medicine, vol.29, No. 6, pp.499-507, ISSN 0785-3890.
Ivanenkov, Y.A., Balakin, K.V., Tkachenko, S.E. (2008). New Approaches to the Treatment of
Inflammatory Disease Focus on Small-Molecule Inhibitors of Signal Transduction
Pathways, Drugs in R&D, vol.9, No. 6, pp.397-434, ISSN 1174-5886.
Jawed, S., Gaffney, K., Blake, D.R. (1997). Intra-articular pressure profile of the knee joint in
a spectrum of inflammatory arthropathies, Annals of the Rheumatic Diseases, vol.56,
No. 11, pp.686-9, ISSN 0003-4967.
Kallberg, H., Padyukov, L., Plenge, R.M., Ronnelid, J., Gregersen, P.K., van der Helm-van
Mil, A., et al. (2007). Gene-Gene and Gene-Environment Interactions Involving
HLA-DRB1, PTPN22, and Smoking in Two Subsets of Rheumatoid Arthritis, The
American Journal of Human Genetics, vol.80, No. 5, pp.867-75, ISSN 0002-9297.
Kleemann, R., Verschuren, L., van Erk, M.J., Nikolsky, Y., Cnubben, N.H.P., Verheij, E.R., et
al. (2007). Atherosclerosis and liver inflammation induced by increased dietary
cholesterol intake: a combined transcriptomics and metabolomics analysis, Genome
Biology, vol.8, No. 9, pp.R200, ISSN 1474-760X.
Lamers, R.J.A.N., DeGroot, J., Spies-Faber, E.J., Jellema, R.H., Kraus, V.B., Verzijl, N., et al.
(2003). Identification of disease- and nutrient-related metabolic fingerprints in
osteoarthritic guinea pigs, Journal of Nutrition, vol.133, No. 6, pp.1776-80, ISSN
1096-0007.
Lassmann, H. (2003). Hypoxia-like tissue injury as a component of multiple sclerosis lesions,
Journal of the Neurological Sciences, vol.206, No. 2, pp.187-91, ISSN 0022-510X.
Lauridsen, M.B., Bliddal, H., Christensen, R., Danneskiold-Samsoe, B., Bennett, R., Keun, H.,
et al. (2010). (1)H NMR Spectroscopy-Based Interventional Metabolic Phenotyping:
A Cohort Study of Rheumatoid Arthritis Patients, Journal of Proteome Research,
vol.9, No. 9, pp.4545-53, ISSN 1535-3907.
Leo, G.C., Darrow, A.L. (2009). NMR-based metabolomics of urine for the atherosclerotic
mouse model using apolipoprotein-E deficient mice, Magnetic Resonance in
Chemistry, vol.47 Suppl 1, pp.S20-S5, ISSN 1097-458X.
Lin, H.M., Edmunds, S.J., Helsby, N.A., Ferguson, L.R., Rowan, D.D. (2009). Nontargeted
Urinary Metabolite Profiling of a Mouse Model of Crohn's Disease, Journal of
Proteome Research, vol.8, No. 4, pp.2045-57, ISSN 1535-3893.
Loffler, M., Zieker, D., Weinreich, J., Lob, S., Konigsrainer, I., Symons, S., et al. (2011).
Wound fluid lactate concentration: a helpful marker for diagnosing soft-tissue
infection in diabetic foot ulcers? Preliminary findings, Diabetic Medicine, vol.28, No.
2, pp.175-8, ISSN 1464-5491.
Lund-Olesen, K. (1970). Oxygen tension in synovial fluids, Arthritis and Rheumatism, vol.13,
No. 6, pp.769-76, ISSN 0004-3591.
Lutz, N.W., Viola, A., Malikova, I., Confort-Gouny, S., Ranjeva, J.P., Pelletier, J., Cozzone,
P.J. (2007). A branched-chain organic acid linked to multiple sclerosis: First
identification by NMR spectroscopy of CSF, Biochemical and Biophysical Research
Communications, vol.354, No. 1, pp.160-4, ISSN 0006-291X.
Metabolomics in the Analysis of Inflammatory Diseases 285

MacGregor, A.J., Snieder, H., Rigby, A.S., Koskenvuo, M., Kaprio, J., Aho, K., Silman, A.J.
(2000). Characterizing the quantitative genetic contribution to rheumatoid arthritis
using data from twins, Arthritis and Rheumatism, vol.43, No. 1, pp.30-7, ISSN 0004-
3591.
Mapp, P.I., Grootveld, M.C., Blake, D.R. (1995). Hypoxia, Oxidative Stress and Rheumatoid-
Arthritis, British Medical Bulletin, vol.51, No. 2, pp.419-36, ISSN 0007-1420.
Marchesi, J.R., Holmes, E., Khan, F., Kochhar, S., Scanlan, P., Shanahan, F., Wilson, I.D.,
Wang, Y.L. (2007). Rapid and noninvasive metabonomic characterization of
inflammatory bowel disease, Journal of Proteome Research, vol.6, No. 2, pp.546-51,
ISSN 1535-3893.
Mayr, M., Madhu, B., Xu, Q. (2007). Proteomics and metabolomics combined in
cardiovascular research, Trends in Cardiovascular Medicine, vol.17, No. 2, pp.43-8,
ISSN 1873-2615.
Metsios, G.S., Stavropoulos-Kalinoglou, A., Panoulas, V.F., Sandoo, A., Toms, T.E., Nevill,
A.M., Koutedakis, Y., Kitas, G.D. (2009). Rheumatoid cachexia and cardiovascular
disease, Clinical and Experimental Rheumatology, vol.27, No. 6, pp.985-8, ISSN 0392-
856X.
Montecucco, F., Mach, F. (2009). Common inflammatory mediators orchestrate
pathophysiological processes in rheumatoid arthritis and atherosclerosis,
Rheumatology, vol.48, No. 1, pp.11-22, ISSN 1462-0324.
Munro, R., Capell, H. (1997). Prevalence of low body mass in rheumatoid arthritis:
Association with the acute phase response, Annals of the Rheumatic Diseases, vol.56,
No. 5, pp.326-9, ISSN 0003-4967.
Murata, Y., Ohteki, T., Koyasu, S., Hamuro, J. (2002). IFN-gamma and pro-inflammatory
cytokine production by antigen-presenting cells is dictated by intracellular thiol
redox status regulated by oxygen tension, European Journal of Immunology, vol.32,
No. 10, pp.2866-73, ISSN 0014-2980.
Murdoch, C., Muthana, M., Lewis, C.E. (2005). Hypoxia regulates macrophage functions in
inflammation, Journal of Immunology, vol.175, No. 10, pp.6257-63, ISSN 0022-1767.
Murdoch, T.B., Fu, H., MacFarlane, S., Sydora, B.C., Fedorak, R.N., Slupsky, C.M. (2008).
Urinary metabolic profiles of inflammatory bowel disease in interleukin-10 gene-
deficient mice, Analytical Chemistry, vol.80, No. 14, pp.5524-31, ISSN 0003-2700.
Naughton, D., Whelan, M., Smith, E.C., Williams, R., Blake, D.R., Grootveld, M. (1993). An
Investigation of the Abnormal Metabolic Status of Synovial-Fluid from Patients
with Rheumatoid-Arthritis by High-Field Proton Nuclear-Magnetic-Resonance
Spectroscopy, FEBS Letters, vol.317, No. 1-2, pp.135-8, ISSN 0014-5793.
Naughton, D.P., Haywood, R., Blake, D.R., Edmonds, S., Hawkes, G.E., Grootveld, M.
(1993). A Comparative-Evaluation of the Metabolic Profiles of Normal and
Inflammatory Knee-Joint Synovial-Fluids by High-Resolution Proton Nmr-
Spectroscopy, FEBS Letters, vol.332, No. 3, pp.221-5, ISSN 0014-5793.
Ng, C.T., Biniecka, M., Kennedy, A., McCormick, J., FitzGerald, O., Bresnihan, B., et al.
(2010). Synovial tissue hypoxia and inflammation in vivo, Annals of the Rheumatic
Diseases, vol.69, No. 7, pp.1389-95, ISSN 0003-4967.
286 Metabolomics

Ngumah, Q.C., Buchthal, S.D., Dacheux, R.F. (2006). Longitudinal non-invasive proton NMR
spectroscopy measurement of vitreous lactate in a rabbit model of ocular
hypertension, Experimental Eye Research, vol.83, No. 2, pp.390-400, ISSN 1096-0007.
Nicoli, F., VionDury, J., ConfortGouny, S., Maillet, S., Gastaut, J.L., Cozzone, P.J. (1996).
Cerebrospinal fluid metabolic profiles in multiple sclerosis and degenerative
dementias obtained by high resolution proton magnetic resonance spectroscopy,
Comptes Rendus de l Academie des Sciences Serie Iii-Sciences de la Vie-Life Sciences,
vol.319, No. 7, pp.623-31, ISSN 0764-4469.
Nishimoto, N., Sugino, H., Lee, H.M. (2010). DNA microarray analysis of rheumatoid
arthritis susceptibility genes identified by genome-wide association studies (vol 12,
pg 403, 2010), Arthritis Research and Therapy, vol.12, No. 3, ISSN 1478-6362.
Parkes, H.G., Grootveld, M.C., Henderson, E.B., Farrell, A., Blake, D.R. (1991). Oxidative
Damage to Synovial-Fluid from the Inflamed Rheumatoid Joint Detected by H-1-
Nmr Spectroscopy, Journal of Pharmaceutical and Biomedical Analysis, vol.9, No. 1,
pp.75-82, ISSN 0731-7085.
Renz, H., von Mutius, E., Brandtzaeg, P., Cookson, W.O., Autenrieth, I.B., Haller, D. (2011).
Gene-environment interactions in chronic inflammatory disease, Nature
Immunology, vol.12, No. 4, pp.273-7, ISSN 1529-2908.
Rieckmann, P., Smith, K.J. (2001). Multiple sclerosis: more than inflammation and
demyelination, Trends in Neurosciences, vol.24, No. 8, pp.435-7, ISSN 0166-2236.
Ross, R. (1999). Mechanisms of disease - Atherosclerosis - An inflammatory disease, New
England Journal of Medicine, vol.340, No. 2, pp.115-26, ISSN 0028-4793.
Scannell, G. (1996). Leukocyte responses to hypoxic/ischemic conditions, New Horizons,
vol.4, No. 2, pp.179-83, ISSN 1063-7389.
Schicho, R., Nazyrova, A., Shaykhutdinov, R., Duggan, G., Vogel, H.J., Storr, M. (2010).
Quantitative metabolomic profiling of serum and urine in DSS-induced ulcerative
colitis of mice by (1)H NMR spectroscopy, Journal of Proteome Research, vol.9, No.
12, pp.6265-73, ISSN 1535-3907.
Serhan, C.N. (2009). Systems approach to inflammation resolution: identification of novel
anti-inflammatory and pro-resolving mediators, Journal of Thrombosis and
Haemostasis, vol.7, pp.44-8, ISSN 1538-7933.
Serkova, N.J., Van Rheen, Z., Tobias, M., Pitzer, J.E., Wilkinson, J.E., Stringer, K.A. (2008).
Utility of magnetic resonance imaging and nuclear magnetic resonance-based
metabolomics for quantification of inflammatory lung injury, American Journal Of
Physiology-Lung Cellular And Molecular Physiology, vol.295, No. 1, pp.L152-L61,
ISSN 1040-0605.
Simone, I.L., Federico, F., Trojano, M., Tortorella, C., Liguori, M., Giannini, P., Picciola, E.,
Natile, G., Livrea, P. (1996). High resolution proton MR spectroscopy of
cerebrospinal fluid in MS patients. Comparison with biochemical changes in
demyelinating plaques, Journal of the Neurological Sciences, vol.144, No. 1-2, pp.182-
90, ISSN 0022-510X.
Sinclair, A.B., Viant, M.R., Ball, A.K., Burdon, M.A., Walker, E.A., Stewart, P.M., Rauz, S.,
Young, S.P. (2010). NMR-Based Metabolomic Analysis of Cerebrospinal Fluid and
Serum in Neurological Diseases - A Diagnostic Tool?, NMR in Biomedicine, vol.23,
No. 2, pp.123-32, ISSN 1099-1492.
Metabolomics in the Analysis of Inflammatory Diseases 287

Sinclair, A.J., Ball, A.K., Burdon, M.A., Clarke, C.E., Stewart, P.M., Cumow, S.J., Rauz, S.
(2008). Exploring the pathogenesis of IIH: An inflammatory perspective, Journal of
Neuroimmunology, vol.201, pp.212-20, ISSN 0165-5728.
Sitkovsky, M., Lukashev, D. (2005). Regulation of immune cells by local. tissue oxygen
tension: Hif1 alpha and adenosine receptors, Nature Reviews Immunology, vol.5, No.
9, pp.712-21, ISSN 1474-1733.
Stappenbeck, T.S., Cadwell, K., Patel, K.K., Maloney, N.S., Liu, T.C., Ng, A.C.Y., et al. (2010).
Virus-Plus-Susceptibility Gene Interaction Determines Crohn's Disease Gene
Atg16L1 Phenotypes in Intestine, Cell, vol.141, No. 7, pp.1135-64, ISSN 0092-8674.
Strober, W., Fuss, I., Mannon, P. (2007). The fundamental basis of inflammatory bowel
disease, Journal of Clinical Investigation, vol.117, No. 3, pp.514-21, ISSN 0021-9738.
Summers, G.D., Deighton, C.M., Rennie, M.J., Booth, A.H. (2008). Rheumatoid cachexia: a
clinical perspective, Rheumatology, vol.47, No. 8, pp.1124-31, ISSN 1462-0324.
Summers, G.D., Metsios, G.S., Stavropoulos-Kalinoglou, A., Kitas, G.D. (2010). Rheumatoid
cachexia and cardiovascular disease, Nature Reviews Rheumatology, vol.6, No. 8,
pp.445-51, ISSN 1759-4790.
Trabold, O., Wagner, S., Wicke, C., Scheuenstuhl, H., Hussain, M.Z., Rosen, N., Seremetiev,
A., Becker, H.D., Hunt, T.K. (2003). Lactate and oxygen constitute a fundamental
regulatory mechanism in wound healing, Wound Repair and Regeneration, vol.11,
No. 6, pp.504-9, ISSN 1524-475X.
Trapp, B.D., Bo, L., Mork, S., Chang, A. (1999). Pathogenesis of tissue injury in MS lesions,
Journal of Neuroimmunology, vol.98, No. 1, pp.49-56, ISSN 0165-5728.
Treuhaft, P.S., McCarty, D.J. (1971). Synovial fluid pH, lactate, oxygen and carbon dioxide
partial pressure in various joint diseases, Arthritis and Rheumatism, vol.14, No. 4,
pp.475-84, ISSN 0004-3591.
Vergadi, E., Chang, M.S., Lee, C., Liang, O.D., Liu, X., Fernandez-Gonzalez, A., Mitsialis,
S.A., Kourembanas, S. (2011). Early macrophage recruitment and alternative
activation are critical for the later development of hypoxia-induced pulmonary
hypertension, Circulation, vol.123, No. 18, pp.1986-95, ISSN 0009-7322.
Wang, Z.N., Klipfell, E., Bennett, B.J., Koeth, R., Levison, B.S., Dugar, B., et al. (2011). Gut
flora metabolism of phosphatidylcholine promotes cardiovascular disease, Nature,
vol.472, No. 7341, pp.57-65, ISSN 0028-0836.
Weljie, A.M., Dowlatabadi, R., Miller, B.J., Vogel, H.J., Jirik, F.R. (2007). An inflammatory
arthritis-associated metabolite biomarker pattern revealed by H-1 NMR
Spectroscopy, Journal of Proteome Research, vol.6, No. 9, pp.3456-64, ISSN 1535-3893.
Wheaton, W.W., Chandel, N.S. (2011). Hypoxia. 2. Hypoxia regulates cellular metabolism,
American Journal of Physiology - Cell Physiology, vol.300, No. 3, pp.C385-C93, ISSN
0363-6143.
White, J.R., Harris, R.A., Lee, S.R., Craigon, M.H., Binley, K., Price, T., Beard, G.L., Mundy,
C.R., Naylor, S. (2004). Genetic amplification of the transcriptional response to
hypoxia as a novel means of identifying regulators of angiogenesis, Genomics,
vol.83, No. 1, pp.1-8, ISSN 0888-7543.
Williams, H.R.T., Cox, I.J., Walker, D.G., North, B.V., Patel, V.M., Marshall, S.E., et al. (2009).
Characterization of Inflammatory Bowel Disease With Urinary Metabolic Profiling,
American Journal of Gastroenterology, vol.104, No. 6, pp.1435-44, ISSN 0002-9270.
288 Metabolomics

Young, S.P., Nessim, M., Falciani, F., Trevino, V., Banerjee, S.P., Scott, R.A.H., Murray, P.I.,
Wallace, G.R. (2009). Metabolomic analysis of human vitreous humor differentiates
ocular inflammatory disease, Molecular Vision, vol.15, No. 125-29, pp.1210-7, ISSN
1090-0535.
Young, S.P., Wallace, G.R. (2009). Metabolomic analysis of human disease and its
application to the eye, Journal of Ocular Biology, Disease and Informatics, vol.2, No. 4,
pp.235-42, ISSN 1936-8445.
Zhai, G., Wang-Sattler, R., Hart, D.J., Arden, N.K., Hakim, A.J., Illig, T., Spector, T.D. (2010).
Serum branched-chain amino acid to histidine ratio: a novel metabolomic
biomarker of knee osteoarthritis, Annals of the Rheumatic Diseases, vol.69, No. 6,
pp.1227-31, ISSN 0003-4967.
12

Clinical Implementation of Metabolomics


Akira Imaizumi et al.*
Amino Acids Basic And Applied Research Group, Frontier Research Laboratories,
Institute for Innovation, Ajinomoto, Co., Inc.
Japan

1. Introduction
1.1 Overview
Metabolomics, which is also referred to as metabonomics, metabolic profiling or metabolic
fingerprinting, is the comprehensive quantitative measurement of endogenous metabolites
within a biological system (Fiehn, 2002; Kaddurah-Daouk et al, 2008; Spratlin et al, 2009).
Detection of metabolites is in general carried out in cell extracts, tissue specimens, or various
biological fluids including serum, plasma, urine and cerebrospinal fluid (CSF) by liquid
chromatography mass spectrometry (LC-MS), gas chromatography–mass spectrometry (GC-
MS), capillary electrophoresis–mass spectrometry (CE-MS) or nuclear magnetic resonance
spectroscopy (NMR). Metabolomics captures the status of diverse biochemical pathways in
a particular situation and can define the metabolic status of an organism (Aranibar et al,
2011; DeFeo et al, 2011; Lu et al, 2008; Roux et al, 2011; Soga, 2007; Yuan et al, 2007). In
clinical settings, biomarkers generated from metabolomics have become one of the most
essential diagnostic criteria that can be objectively measured and evaluated as indicators of
normal or pathological states, as well as a tool to assess responses to therapeutic
interventions (Hunter, 2009; Spratlin et al, 2009; van der Greef et al, 2006; Zeisel, 2007). As
we describe in this chapter, novel metabolomic markers, for instance, for cancer therapy,
glucose intolerance, hepatic steatosis, nephrotic and psychiatric disorders, and their
incorporation into clinical decision-making may considerably change future health care.
In order for metabolomics to be successful in clinical settings, it must surpass
conventional methods in reliability and predictive capability, and/or should be more
informative about disease pathogenesis. Utilizing a systems biology approach in

*Natsumi Nishikata1, Hiroo Yoshida2, Junya Yoneda1, Shunji Takahena2,

Mitsuo Takahashi1, Toshihiko Ando3, Hiroshi Miyano2, Kenji Nagao1, Yasushi Noguchi1,
Nobuhisa Shimba3,4 and Takeshi Kimura4
1Amino Acids Basic And Applied Research Group, Frontier Research Laboratories,

Institute for Innovation, Ajinomoto, Co., Inc., Japan


2Analytical Sciences Group, Fundamental Technology Laboratories, Japan

Institute for Innovation, Ajinomoto, Co., Inc.


3AminoIndex Department, Wellness Business Division, Ajinomoto, Co., Inc., Japan

4R&D Planning Department, Ajinomoto, Co., Inc., Japan


290 Metabolomics

biomarker investigation may allow for a deeper understanding of disease associated


metabolism. (Jenkins et al, 2004; Kell, 2006). A systems biology approach does not focus
on identifying a single target or mechanism of an observed phenotype. Instead it seeks to
identify the biological networks or pathways that connect the differing elements of a
system (Wheelock et al, 2009). When a shift in equilibrium is observed in a disease, such
as altered metabolic fluxes or enzymatic activities, it can be elucidated that those
components of the network that are associated with the observed shift are characteristic
and potentially descriptive of the disease, and that they accordingly represent potential
targets for intervention. Thus, the systems approach in combination with metabolomics,
may lead to the discovery of panels of metabolites that more accurately capture the
disease status and help acquire information valuable for individualized clinical care
(Quinones & Kaddurah-Daouk, 2009). Clinical metabolomics is expected to be a
promising technology for personalized medicine and nutrition. A metabolic marker
designed to predict individual response including efficacy and side effects during
therapeutic intervention for each patient will enable administration of optimal treatments
and improve clinical outcome.

1.2 Comprehensive vs. focused metabolomics


The spectrum of biochemicals in a clinical specimen, range from organic acids, amino acids,
lipids, nucleic acids and their metabolic intermediates to complex secondary metabolites
with signaling functions. Today, however, clinicians in human health care utilize only a very
small part of the information contained in the metabolome. Although NMR or MS
technology enables a comprehensive (i.e. global) measurement of various small molecules,
in many cases, it is simply too difficult to quantify each molecule and understand
underlying mechanisms from a global dataset by a single measurement (Steuer, 2006). This
has led a number of researchers to look at a focused set of (i.e. local) metabolites such as
amino acids or lipids (German et al, 2007; Kimura et al, 2009), where data from multiple
measurements such as transcriptomics, proteomics and metabolomics can be effectively
integrated to allow more insight into the underlying metabolic alternations, by projecting
multiple datasets onto biochemical pathways and analyzing their interactions under a
particular physiological state (Caesar et al, 2010; Momin et al, 2011; Noguchi et al, 2008;
Zhang et al, 2011).
Recently, there have been reports of trials in integrating different types of ‘omics’ datasets
for the systemic understanding of metabolic phenotypes at multiple levels. Various
software packages are available in integrating nonuniform ‘omics’ datasets (Grimplet et
al, 2009; Gruning et al, 2010; Taylor & Singhal, 2009). The link between information and
modeling can be achieved by two major types of complementary approaches, a data-
oriented exploratory approach, in which data generates information about the structure
and relationships between the observed variables in a given system, and a model-based
bottom-up approach, in which cybernetic and systems–theoretical knowledge are used to
create models that describe mechanisms and dynamics of a system. Formerly, a model-
based approach had been used for studying in-vitro cellular or organ systems; however,
because of the complexity in modeling whole body systems, recently this approach has
been replaced by a data-oriented approach, particularly when dealing with in-vivo
‘omics’ data in various models including animal models and clinical studies (Dunn et al,
Clinical Implementation of Metabolomics 291

2011). The critical step is the construction of models from the raw dataset of
transcriptomics, proteomics, and metabolomics. This may be achieved by using different
mathematical techniques ranging from simple Pearson correlations to the use of ordinary
differential equations (Wheelock et al, 2009). Through this modeling, fundamental
concepts in the understanding of biological systems like robustness, modularity,
emergence, etc. are incorporated.
Most studies currently remain focused on local level networks within a set of related genes
or protein expressions (Bapat et al, 2010; Kirouac et al, 2010). Yet a combination of different
levels of networks can be connected to overview the whole system. A change in the gene
regulatory network may have a corresponding effect in the protein–protein interaction
network, the metabolic network, etc., which collectively may manifest changes in the
pathological phenotype. To understand the whole system, it is critical to integrate
knowledge from different datasets. Although some progress has been made in amino acid
metabolism, the integration of different types of datasets is still difficult due to differences in
dynamic range, scales, or analytical errors, particularly in metabolomic analysis (Ishii et al,
2007; Momin et al, 2011; Noguchi et al, 2008). Therefore, focused-metabolomics, with well
managed measurements in terms of accuracy and reproducibility, for lipid, amino acid and
glucose metabolism appears to be a realistic approach to illustrate how the phenotype is
altered when the metabolic network itself is modified through the alteration of endogenous
or environmental factors.

1.3 Generation of multiple metabolite markers


When generating biomarkers from metabolomic analysis, marker identification, verification,
and also statistical and experimental evaluations, using bioinformatic techniques of
identified candidate markers are required. Recently, various data mining methodologies
have been reported for identifying and prioritizing reliable metabolomic markers with high
diagnostic capability (Caruana, 2006; Duda, 2001; Gu et al, 2011; Kim et al, 2010; Maeda et al,
2010; Montoliu et al, 2009). In cohort studies, the definite diagnoses of the patients are
normally known beforehand. In such trials, “supervised” statistical methods which consider
patient classification tend to be more efficient in information utilization and suitable for
obtaining targeted metabolite markers.. In contrast, when phenotypes in patients are
undetermined, “unsupervised” analysis such as cluster analysis are useful tools for
biomarker identification and classification of specimen groups. Moreover, improvement in
discriminatory power has been reported when multivariate mathematical models are
constructed combining multiple metabolite markers. These approaches include discriminant
analysis methods such as linear discriminant analysis, logistic regression analysis, decision
trees, the k-nearest neighbor classifier (k-NN), an instance-based learning algorithm,
support vector machines or artificial neural networks (Duda, 2001). The Receiver Operating
Characteristics (ROC), or the area under the ROC curve (AUC) of multivariate markers is
used to represent its discriminatory performance as a trade off between selectivity and
sensitivity(Hanley & McNeil, 1982). Obtained metabolomic markers are also required to be
experimentally validated using larger datasets from multiple clinical trials and also
statistically validated using cross validation, leave-one-out cross validation, and
bootstrapping.
292 Metabolomics

2. Practical Issues in the clinical implementation of metabolomics


2.1 Sample stability issues
Enormous information can be obtained by analyzing large numbers of metabolites, and it is
utilized for various fields such as health and nutrition. However, the chemical and
enzymatic stabilities of most metabolites are unknown. Therefore, inappropriate handling of
samples can lead to inaccurate measurements. In this section, blood sampling issues for
amino acids analysis as a typical case of sample handling are described. There are mainly
four steps in the blood sampling process for amino acids analysis; 1) blood collection, 2)
centrifugation, 3) sample storage, and 4) deproteinization. In this section, the crucial points
for each step are outlined to highlight the importance of sampling processes in metabolomic
studies.

2.1.1 Blood collection


The concentrations of amino acids are known to show circadian rhythms and some of
them vary 30% within a day (Forslund et al, 2000). Therefore, it is desirable to collect the
blood at a fixed time point. Moreover, since the amino acid concentrations increase after a
protein containing meal, blood collecting between 7am and 10am in a fasting state is
desirable.
The concentrations of some amino acids are known to be quite different between blood cells
and plasma. The differences of essential amino acids are small, but the concentrations of
nonessential amino acids can be greater by severalfold in blood cells (Filho et al, 1997).
There are also many metabolic enzymes such as arginase in blood cells which will act on the
plasma free amino acids (PFAAs). Therefore it is important to verify that haemolysis dose
not occur in blood samples. If the blood sample shows heavy haemolysis, it is desirable to
take another sample.
If blood samples are left at room temperature after collection until centrifugation, many
amino acids are metabolized due to metabolic enzymes from blood cells. In particular, there
are many enzymes for metabolizing nonessential amino acids. For instance, glutamine and
asparagine are well known to be metabolized to glutamate and aspartate. The concentration
change of glutamate at different temperatures is shown in Figure 1. This suggests that it is
desirable to cool blood samples after collecting. In another study, we also found that it is
essential to cool down the blood samples to 0°C immediately after collecting and that Ice-
water is better than the refrigerator or ice because of the faster cooling rate.
However it is not always easy to prepare ice-water in the medical institutions at the time of
blood collection. For this reason, we have developed a portable blood tube cooler
(CubeCooler™, Figure 2). This cooler is composed of high thermal conductive container
(aluminum) and insulator (polyethylene form), which enables the quick cooling of blood
samples as well as ice-water and maintains the temperature for 12h (Figure 3). There are
many coolers which is commercially available. As far as we have examined, these coolers,
however, could not achieve a cooling rate as close to that of ice-water and could not cool
blood samples for a long time without differences in temperature arising between tubes
inserted in different holes. Thus, the cooler we have developed may be a useful tool not only
for amino acid analysis but also for sample management in other metabolomic studies.
Clinical Implementation of Metabolomics 293

1.7 RT

Relative Conc.
1.6 Fridge
1.5 Ice-Water
1.4
1.3
1.2
1.1
1

0 2 4 6
Time(h)
Fig. 1. Effect of cooling on concentration of glutamate in whole blood

Fig. 2. View of the blood tube cooler (CubeCooler™)

40
35
Room Temp.
30
25
Fridge
20 Ice
Temp. (°C )

15
10
Blood Tube Cooler
5 Blood Tube Cooler
Ice-Water
0
-5
0 3 6 9 12 15 2 6 10 14
Time (min) Time (h)

Fig. 3. Cooling rate when the blood tubes are set in various conditions and cooling duration
of the blood tube cooler
294 Metabolomics

2.1.2 Centrifugation
It is desirable to store blood samples in ice-water after collection and to separate the plasma
from the blood cells within a few hours. As mentioned above, since blood cells contains
many amino acids and enzymes, it is important not to contaminate the plasma with
platelets. If contamination occurs, the concentrations of some amino acids, such as
glutamate, aspartic acid and taurine can be high.

2.1.3 Sample storage


It is necessary to store the plasma in a freezer in case of long term storage. When stored at -
20°C, some amino acids, especially glutamate, aspartate and cysteine can gradually
decrease. Therefore -80°C freezer should be used for long term storage of plasma samples.
When transporting the samples, the samples should be carried in a box filled with dry-ice.

2.1.4 Deproteinization
Since plasma contains proteins such as albumin, deproteinization is necessary before amino
acid analysis. When analyzed with amino acid analyzer, plasma is generally mixed with
trichloro-acetic acid or sulfo-salicylic acid and the precipitate is centrifuged. Since these
reagents are strong acids, it is necessary to rapidly analyze amino acids or store in -80°C
freezer so that some amino acids like glutamine are not decomposed due to acid hydrolysis.
When analyzing with LC-MS or LC-MS/MS, organic solvents such as methanol and
acetonitrile is useful for deproteinization. In this case, the organic solvent may influence the
derivatization reaction and separation of amino acids. Since recovery rates for amino acids
depend on the procedure of deproteinization, it is desirable to unify the procedure. When
analyzing with LC-MS or LC-MS/MS, recovery rates can be calculated by adding stable-
isotope-labeled amino acids as internal standards before deproteinization.

2.2 Analytical issues


Nuclear magnetic resonance (Bollard et al, 2001), mass spectrometry (Piraud et al, 2003), gas
chromatography mass spectrometry (Thysell et al, 2010), liquid chromatography mass
spectrometry (LC-MS) (Lin et al, 2011a), and capillary electrophoresis mass spectrometry
(Sugimoto et al, 2010) have been used as primary tools employed for metabolomics.
A clinical metabolomics approach with LC-MS can be broadly classified into comprehensive
and targeted analysis. Comprehensive analysis aims to identify and quantify all detectable
metabolites in a single run. This analysis offers the advantage of giving much information.
In the past, the retention and separation of polar metabolites had been difficult in LC-MS
analysis. This was a weakness of LC-MS analysis, and LC-MS was limited to the analysis of
hydrophobic metabolites such as lipids. However, the development of column technology
enabled the retention and separation of hydrophilic metabolites(Alpert, 1990; Yoshida et al,
2007). This technology has been applied for the research of drug metabolites (Plumb et al,
2003), galactosamine toxicity (Spagou et al, 2011), and renal cell carcinoma diagnosis,
staging, and biomarker discovery (Lin et al, 2011a).
In targeted analysis, a selected number of predefined metabolites are quantified. This analysis
is sometimes used for quantification of metabolites, which is extracted from comprehensive
Clinical Implementation of Metabolomics 295

analysis. Derivatization methods, based on specific reactions to targeted functional groups are
major tools in targeted analysis. This method allows for sensitive and selective quantification
of endogenous metabolites with amino and carboxyl groups (Tsukamoto et al, 2006; Yang et al,
2006). An advantage of this method is to be able to select a suitable sample preparation for
each endogenous metabolite with the same functional group, because of the similar physical
and chemical properties. This method is also very important for accurate quantification,
because sample stability is different for each endogenous metabolite.
The analysis of amino acids with an amino group has a long history. In 1958, a key
application for physiological amino acid analysis was supplanted by ion exchange column
chromatography separations on an automated apparatus designed and built by postdoctoral
fellow Darrel H. Spackman at the request of his mentor William H. Stein, and Stanford
Moore at Rockerfeller University (Moore et al, 1958). This automated system reduced the
analytical time from a few weeks to a full day and provided easy to use operation. The
present system is used for the study of inborn errors of amino acid metabolism in clinical
laboratories (Qu et al, 2001).
Recently, pre-column derivatization reagents for amino acid analyses have been developed,
mainly to achieve greater sensitivity and selectivity, and much attention is paid to the
design of derivatization reagents for LC-MS (Yang et al, 2006) and LC-MS/MS (Shimbo et al,
2009a; Shimbo et al, 2009b). These reagents have three notable characteristics (Figure 4).
First, the reagent must have sufficient hydrophobicity to enable the retention of amino acids.
Secondly, is should have a desirable structure which will increases ionization efficiency.
Thirdly, it should be designed to provide characteristic and selective cleavage at the
bonding site between the reagent moiety and the amino acid in the collision cell of the triple-
stage quadrupole mass spectrometer. Using precursor ion scanning, endogenous
metabolites with amino groups are can be extracted on ion chromatograms, even in crude
biological samples.
3-aminopyridyl-N-hydroxysuccinimidyl carbamate (APDS) reagent is known to provide
rapid analysis and separation of amino acids of the same charge to mass ratio on a column
(Shimbo et al, 2009b) (Figure 5). This reagent is applied to the modelling of a diagnostic
index, “AminoIndex technology”, from differences in PFAA profiles between non-cachectic
colorectal/breast/lung cancer patients and healthy individuals. (Maeda et al, 2010;
Okamoto et al, 2009).

Fig. 4. Typical reaction of amino acids with a derivatizaiton reagent for LC-MS/MS. This
reagent has three notable characteristics; 1) sufficient hydrophobicity (benzene ring) 2)
increases ionization efficiency (quaternary amine) 3) characteristic and selective cleavage
(the reagent moiety and the amino acid).
296 Metabolomics

Fig. 5. Typical chromatograms of amino acids which were the same charge to mass ratio on
a column.

2.3 Statistical issues


Recently, several applications of metabolome analysis based on computer-aided detection
and diagnosis (CAD) has been demonstrated (Duda, 2001; Gu et al, 2011; Kell, 2002; Kim et
al, 2010; Montoliu et al, 2009; Righi et al, 2009; Serkova et al, 2011; Taylor et al, 2010). The
importance of objective indices for diagnosis based on empirical and statistical knowledge
are increasing due to the trend called “Evidence Based Medicine (EBM)”. Although CAD
follows this trend, the required level of statistical analysis is also increasing and becoming
more complex. The requirement of clinical investigation includes not only statistical
significance but also feasible and in-depth clinical protocols in which necessary and
sufficient conditions need to be satisfied. In this section, multivariate statistical aspects of
metabolome analysis focused on the establishment of medical evidence and investigation of
biomarkers will be introduced and discussed.
Reproducibility is the most important point of a diagnostics index. It is more complicated to
guarantee the statistical reproducibility by multivariate analysis than univariate analysis.
Adequate experimental design prior to data collection is therefore crucial for the quality
control of the analysis (Hulley, 2006). In general, knowledge obtained from statistical
analysis is only capable within the realm in which the data was analyzed and therefore
cannot extrapolate beyond the realm. Generally, larger sample size is required in case of
multivariate analysis because freedom of variable space is higher than univariate analysis.
For example, multivariate analysis of variance (MANOVA) and data simulation are used to
determine the appropriate sample size. Additionally, it is sometimes necessary for a data set
to be normalized or scaled for unbiased analysis.
Clinical Implementation of Metabolomics 297

The most important point of analysis is algorithm selection. It is well-known as the “no free-
lunch theorem”, that it is impossible to determine the most suitable algorithm a priori, and
that the pros and cons of each algorithm are not always specific, but dependent on each
situation. Therefore, preliminary analysis to determine the most felicitous algorithm is
necessary in each case. Univariate analysis can be performed to figure the behavior of each
metabolite and to select the variable, i.e. dimensionality reduction of variable space, prior to
multivariate analysis. It should be noted that the metabolome data are often so connected
that there is a potential pitfall of statistical analysis, so-called multicollinearity, where the
excess reduction of dimension sometimes can lead to the loss of latent network structure of
metabolites. Multivariate analytical methods are applicable for simplification or
dimensionality reduction of data to easily figure out visualized images of the “metabolite
space” which has huge body of dimensions (metabolites).
Algorithms for multivariate analyses are categorized into two different groups, i.e.,
unsupervised methods and supervised methods. Unsupervised methods do not require
objective variables such as subject status, other observed data, etc., while supervised
methods require them for the data set to be analyzed. The examples of multivariate
algorithms are listed in Table 1. Unsupervised learning methods are especially useful for
investigating the latent structure and decreasing the redundancy of data and therefore they
are sometime performed in combination. The advantages of unsupervised methods are that
they minimize the loss of information (Maeda et al, 2010). However, whether the results of
unsupervised methods can provide the appropriate interpretation or not depends on the
setting of parameters or the problem to be analyzed.

Models Unsupervised learning Supervised learning


Continuous Discrete
Multiple linear regression Linear discriminant analysis
Linear model Factor analysis
(MLR) (LDA)
Principal component analysis Canonical correlation
Canonical discriminant analysis
(PCA) analysis
Independent component Partial least square regression Partial least square discriminant
analysis (ICA) (PLS) analysis (PLS-DA)
Hierarchical cluster analysis
Nonlinear model Logistic regression analysis Naïve Bayes classifier
(HCA)
Conditional logistic regression
K-means cluster analysis Support vector machine (SVM)
analysis
Generalized linear model
Mixture of Gaussians Decision trees
(GLM)

Table 1. Algorithm examples for multivariate analysis

On the contrary, supervised methods (Caruana, 2006) themselves contain the objective
variables. Therefore the goal of analysis is to find a model (or classifier) in which the error
between the model’s response and the target traits is minimized to fit the target traits. Target
traits can be discrete (e.g., disease vs. healthy, grade of disease) or continuous (e.g.,
measurement value). Supervised methods are also applicable to discover and predict which
metabolites are responsible for the target traits (Maeda et al, 2010; Okamoto et al, 2009;
Zhang et al, 2006). However, the generality of the model obtained from those methods can
not be always guaranteed because of the potential overfitting or bias of data. Therefore,
298 Metabolomics

validation of the obtained model is necessary to establish the usefulness for practical use.
Validation methods are categorized into two classes. The first is cross- validation in which
single or multiple samples are iteratively left out from the training data set, and the
remaining samples are used to evaluate the predictive performance of the model. The other
is usage of external validation data set which must not be used for construction of models.
Ideally, the latter case in which blinded data set is used is the most appropriate validation.
However, it is sometimes difficult to perform the validation test itself.
Various metrics are used as criterion of the performance of diagnosis. In the case of the
model in which the object variable contains only two classes (e.g., controls and patients),
receiver-operator characteristic (ROC) curve analysis is the most appropriate criteria for
evaluating the model because this analysis is independent of both sample size of each group
and threshold. As threshold metrics, sensitivity, specificity, positive predictive value (PPV),
negative predictive value (NPV), and accuracy are used. Among them, both sensitivity and
specificity is independent of sample size and ratio of each group while the others are
dependent. Therefore, to determine threshold in terms of PPV, NPV, and accuracy, it is
necessary to take into account the “real” distribution of subjects.

3. Examples of clinical implementation of focused metabolomics


3.1 „AminoIndex technology“: Example for early cancer diagnosis
Several investigators have reported changes in plasma free amino acid (PFAA) profiles in
cancer patients (Cascino et al, 1995; Lai et al, 2005; Lee et al, 2003; Maeda et al, 2010; Naini et
al, 1988; Norton et al, 1985; Okamoto et al, 2009; Proenza et al, 2003; Vissers et al, 2005;
Zhang & Pang, 1992). Despite evidence of a relationship between PFAA profiles and some
types of cancer, few studies have explored the use of PFAA profiles for diagnosis because
although PFAA profiles differ significantly between patients, the differences in individual
amino acids do not always provide sufficient discrimination abilities by themselves (Cascino
et al, 1995; Lai et al, 2005; Naini et al, 1988; Norton et al, 1985; Proenza et al, 2003; Vissers et
al, 2005). To address this issue, we have studied using diagnostic indices based on PFAA
concentrations that compress multidimensional information from PFAA profiles into a
single dimension to maximize the differences between patients and controls.
In previous studies, the alterations in PFAA profiles in cancer patients sometimes seem
inconsistent, and some discrepancies existed between our study and those reported (Cascino
et al, 1995; Lai et al, 2005; Naini et al, 1988; Norton et al, 1985; Proenza et al, 2003; Vissers et
al, 2005). This discrepancy may be due not only to the statistical aspect of data, for example,
sample size, the biased distribution of cancer stages, etc., but also to some other factors such
as amino acid measurement methods. In contrast to previous studies, we performed
analyses using samples in which PFAAs were measured in a unified protocol to guarantee
the robustness of analysis in terms of the quality of data (Shimbo et al, 2009a; Shimbo et al,
2009b; Shimbo et al, 2009c).
As a pilot study, we investigated the possibility for early detection of colorectal cancer
(CRC) and breast cancer (BC) patients (Okamoto et al, 2009). PFAA profiles were compared
between cancer patients (who had CRC or BC) and control subjects. The plasma
concentrations of several amino acids in the CRC patients were significantly different from
Clinical Implementation of Metabolomics 299

those observed in the controls. The alteration of the PFAA profile in BC differed from that in
CRC, with fewer changes observed. Multiple logistic regression analyses with selected
variables using each data set resulted in AUC of ROC of0.860 for CRC and 0.906 for BC,
respectively when using training data sets. To confirm the performance of the obtained
classifier, ROC curves were also generated from the split test data. These reproduced similar
diagnostic performances, with AUC of 0.910 for CRC, and 0.865 for BC, respectively.
We then investigated the possibility for early detection of non-small-cell lung cancer
(NSCLC) using a larger size of samples (Maeda et al, 2010). 141 NSCLC patients and 423
age-matched, gender-matched healthy controls without apparent cancers were used as the
study data set. As a result, fifteen amino acids (Ser, Gly, Ala, Cit, Val, Met, Ile, Leu, Tyr, Phe,
His, Trp, Orn, Lys, and Arg) were identified whose profile in plasma were associated with
NSCLC. Multiple logistic regression analyses by conditional likelihood methods were
performed with variable selection and LOOCV cross-validation using the study data set.
The resulting conditional logistic regression model included six amino acids: Ala, Val, Ile,
His, Trp, and Orn. The AUC of ROC for the discriminant score was 0.817 in the study data
set. It should be noted that conditional logistic (c-logistic) regression analysis can correct the
effects of age, gender, and smoking statuses which are potential confounding factors in the
discrimination. To verify the robustness of the resulting model, a ROC curve was also
generated using the split test data set, which had not been used to construct the model. An
AUC of ROC for the discriminant score was 0.812 in the test data set, again demonstrating
that the obtained model performed well (Figure 6).

Fig. 6. ROC curves for discriminant scores for the discrimination of NSCLC(Maeda et al,
2010).

It was indicated that the model could discriminate lung cancer patients regardless of cancer
stage or histological type. Furthermore, the distribution of the discriminant scores for small-
cell lung cancer (SCLC) patients was similar to that for NSCLC patients (Figure 7).
300 Metabolomics

Fig. 7. ROC curves for discriminant scores subgrouped by NSCLC stage and histological
type (Maeda et al, 2010). A. ROC curves for cancer stage of study data set. B. ROC curves for
cancer stage of test data set. C. ROC curves for histological type of study data set D. ROC
curves for histological type of test data set (including SCLC patients).

These studies demonstrated the potential use of PFAA profiling as a focused


metabolomics approach for the early detection of patients with various types of cancer.
Combining novel analytical techniques and statistical analyses, previously unknown
aspects of amino acid metabolism in humans have been revealed. The analysis using
considerably larger sample size provided sufficient statistical power to test the
robustness of PFAA profiling for cancer diagnosis. We also demonstrated the possibility
of detecting cancers, both specifically and broadly, using multivariate analysis to
compress the PFAA profile data, even for patients with early stage cancer. Following the
further accumulation of data (not shown), AminoIndex® Cancer Screening (AICS) has
been commercially released from Ajinomoto Co., Inc., in Japan in April 2011. AICS
enables multiple cancer diagnoses simultaneously of gastric, lung, colorectal, prostate
and breast cancer.
Clinical Implementation of Metabolomics 301

3.2 „AminoIndex technology“: Example for diagnosis of liver fibrosis


In the clinical pathway of patients with chronic hepatitis C infection, the progression of liver
fibrosis leads to cirrhosis and eventually increases the risk of hepatocellular carcinoma
(Poynard et al, 2003). The efficacy of current therapy depends on the fibrosis grade, and
therefore the detection of fibrosis stage is desirable for determining the clinical settings, i.e.,
whether treatment is necessary, and what treatment is appropriate (Aspinall & Pockros,
2004; Fried, 2002; Shiffman, 2004). Although fibrosis grading based on biopsy has been
considered as a gold standard, there is a high demand for less invasive but effective
alternative methods.
In searching for surrogate markers other than biopsy, several methods ranging from the
serologic marker-based test (Fibrotest)(Imbert-Bismut et al, 2001) to the ultrasonic-based
transient elastography (Fibroscan)(Castera et al, 2005), and others(Lin et al, 2011b) have been
suggested. On the other hand, since the liver is an important organ for the metabolism of
amino acids, glucose synthesis, fatty acid synthesis, urea synthesis and protein
synthesis(Cynober, 2004), it is reasonable to expect any metabolic derangement due to liver
failure like liver fibrosis may induce the variation of amino acid metabolism and eventually
the variation of PFAA concentration.
In this section we describe the PFAA profiling which was first applied to the diagnosis of
liver fibrosis using clinical data(Zhang et al, 2006). The aim of this study was to develop a
diagnostics index for the diagnosis of liver fibrosis as a less invasive and effective method
using PFAA profiles. The liver specimens were analyzed histologically and graded with the
METAVIR scoring system(Metavir., 1994), where F0 means no fibrosis, F1 portal fibrosis
without septa, F2 fibrosis with rare septa, F3 portal fibrosis with numerous septa, and F4
cirrhosis. The distribution and variation of the 23 PFAAs of all patients over fibrosis stages
is represented in a radar chart, Figure 8.
In the progression of fibrosis from F01 to F4, the decrease of BCAA and inversely the
increase of aromatic amino acids, Phe and Tyr, can be observed typically in the profiles of
the radar chart. In the non-parametric multi-stage comparison test (Kruskal-Wallis test) , for
each amino acid among different fibrosis stages, significant changes in concentration of Phe,
Val, Ile, Tyr, Gln, Leu, Met (p <0.01) and ABA (alpha-amino butyric acid, p <0.05) were
observed. Dataset including fibrosis stage and PFAA concentrations were analyzed to obtain
the diagnostics index for liver fibrosis (AI_fibrosis) in fractional form, (Phe)/(Val) +
(Thr+Met+Orn)/(Pro+Gly), which was optimized as a surrogate marker for the liver stages
obtained through biopsies. The distribution of molar ratios in two fractional forms over
fibrosis stages are shown in Figure 9.
The observation of two molar ratios in the classifier revealed that the former ratio mainly
contributed to the F4 discrimination, whereas the latter mainly contributed to discrimination
of advanced fibrosis (F3 and F4). For the discriminative power assessment of the surrogate
AI_fibrosis as a whole, the area under the curve of receiver operator characteristic curve
(ROC AUC) was used. The classifier exhibited high discriminative power for advanced
fibrosis (fibrosis stages F3 and F4) from the earlier stages F0-2 and also for cirrhosis (F4)
from all other stages, with ROC AUC ( 95% CI ) 0.92 (0.84-1.00 ) and 0.99 ( 0.96-1.00 ),
respectively.
302 Metabolomics

Fig. 8. Radar chart of mean values of PFAAs over fibrosis stages. F01: dashed, F2: dot-dash,
F3: dotted, F4: solid. Mean values are scaled in z-score.

Fig. 9. Molar ratio variation over fibrosis stages. The change in distribution among F0-F2,F3
and F4 stages indicated a stage-dependent trend. Circles are 80% regions of each stage, F0-
F2: dashed and square, F3: dotted and triangle, and F4: solid and christcross.
Clinical Implementation of Metabolomics 303

The Fischer’s ratio (Val+Leu+Ile)/(Phe+Tyr) was originally created for diagnosis of hepatic
encephalopathy (Fischer et al, 1975; Fischer et al, 1976) and has been reported to show good
performance in assessing chronic hepatitis (Kano et al, 1991). Therefore a comparison study
between the Fischer’s ratio and the classifier was undertaken, where the index was
generated to have a positive correlation with the degree of fibrosis, showing an inverse
pattern to Fischer’s ratio. The AI_fibrosis indicated ROC AUC values larger than Fischer’s
ratio: the ROC AUC values of Fischer’s ratio being 0.87 (0.77-0.96) for advanced fibrosis and
0.91 (0.83-0.99) for cirrhosis, respectively. There is a close relationship between the
AI_fibrosis and the Fischer’s ratio as partially supported by the fact that the ratio Phe/Val
correlated well with the inverse of Fischer’s ratio (r = 0.95) because the BCAAs exhibited
good mutual correlation, as did Tyr and Phe. In summary, these results suggest that the
AI_fibrosis based on amino acid concentration can be applied to evaluate liver fibrosis as an
effective and less invasive method as a surrogate marker for liver biopsy, although future
extended validation study is still necessary.

3.3 Lipidomics: A review on the use of lipid metabolomics for clinical use
Lipidomics, a type of focused metabolomics, is the comprehensive measurement of a variety
of lipid classes: free fatty acids (FFA), triglycerides (TAGs), cholesterol esters (CEs),
lysophosphatidylcholines (LPCs), phosphatidylcholines (PCs), lysophosphatidyl
ethanolamines (LPEs), diacylglycerols (DAGs), and sphingomyelins (SMs) and ceramides,
generally using LC-MS/MS (Bou Khalil et al, 2010; Bucci, 2011; Dennis, 2009). Several
studies have reported the potency of lipidomic analyses for biomarker discoveries in
humans in diabetes, non-alcoholic fatty liver disease (NAFLD) (Puri et al, 2009), Alzheimer’s
disease (Han et al, 2011; Valdes-Gonzalez et al, 2011) and cancers (Hilvo et al, 2011; Min et
al, 2011). For instance, Rhee et al reported the LC-MS–based lipid profiling of 189
individuals who developed type 2 diabetes and 189 matched disease-free individuals, with
over 12 years of follow up in the Framingham Heart Study (Rhee et al, 2011). They found
that lipids of lower carbon number and double bond content were associated with an
increased risk of diabetes, whereas lipids of higher carbon number and double bond content
were associated with a decreased risk. In addition, Barr et al demonstrated differential
serum lipidomics in both NAFLD patients and in a mouse model of NAFLD by ultra
performance liquid chromatography-mass spectrometry (UPLC-MS) (Barr et al, 2010).
Multivariate statistical analysis of the UPLC-MS datasets revealed metabolic similarities
between NAFLD mice and human NAFLD patients in relative serum metabolite levels
compared to normal subjects. Lipidomic analysis is also applicable to other biological fluids
such as cerebrospinal fluid (CSF), in addition to plasma and serum (Fonteh et al, 2006). For
instance, phospholipid profiling in the CSF by nano-HPLC-MS has been reported in
Alzheimer’s disease (AD) patients, and a statistically significant increase of SMs were
observed in CSF from probable AD patients compared to normal subjects (Han et al, 2011).

4. Elucidation of mechanisms underlying metabolomic diagnosis


4.1 Introduction
Many living systems have homeostatic mechanisms to continuously maintain their
biological activity. Yet, when a dynamic multi-parametric metabolic response to patho-
304 Metabolomics

physiological stimuli is evoked in many disease-associated cells and tissues, it leads to the
formation of disease-specific enzymatic metabolite profiles quite different from that of the
healthy hosts, and the blood components are significantly influenced as a result.
Blood amino-acid contents are included in such components (referred to as a blood amino-
acid profile). It is well known that in the process of feeding, exercising, sleeping, and other
activities, the blood amino-acid profile temporarily fluctuates, but within a few hours
returns to the normal level through intrinsic homeostatic mechanisms. By contrast, disease-
mediated disturbances in the local amino-acid metabolisms may result in formation of a
disease-specific change in the blood amino-acid profile. Based on these findings and
discussions, we have introduced the AminoIndex® Cancer Screening (AICS) system as a
tool for providing new biomarkers to enable the early detection of various cancers.

4.2 Tumor-specific blood amino-acid profile


In order to explain the effects on PFAA profiles by the various tumors, we propose the
following simple idea consisting of “three components”. As shown in Figure 10, these three
components are as follows: a) Metabolic changes in the tumor-bearing organs; b) Metabolic
changes in response to the inflammatory reactions; and c) Metabolic changes in various
remote healthy organs. In patients with tumors, these three metabolic changes may be
evoked simultaneously and their overall effects may be reflected in the tumor-specific blood
amino-acid profile. Yet, it is highly unlikely that they contribute evenly to such formation of
the tumor-specific blood amino-acid profile during the entire course of the tumor
development. It seems more reasonable that these three components contribute
individually and differently to the formation of the tumor-specific blood amino-acid profile
in the early, the mid and the late (cachexia) stages.

Fig. 10. Scheme for mechanisms underlying tumor-specific metabolic changes

In tumor-bearing hosts, “metabolic changes in tumor-bearing organs”, “metabolic changes


in response to inflammations” and “metabolic changes in remote organs” are all evoked
simultaneously, leading to formation of the tumor-specific amino-acid profile.
Clinical Implementation of Metabolomics 305

4.3 Metabolic changes in the tumor-bearing organs


It has been shown that cancer cells, which can proliferate extraordinarily faster than
healthy normal cells, obtain the biological energy required to proliferate by switching to
aerobic glycolysis from oxidative phosphorylation (Matthew et al, 2009)(known as
“Warburg effect”): several intermediates obtained from the glycolysis pathway might be
more easily utilized in nucleotide synthesis and glucose might be used as a carbon
source in fatty acid generation (Dang, 2010). At the same time, glutaminolysis was
found to be stimulated profoundly, meaning that cancer cell energy generation is highly
dependent on glutamine content (Wise & Thompson, 2010). A change in the amino-acid
metabolism has been documented even in several noncancerous areas of the tumor-
bearing organs. Douvlis has proposed the possibility that many normal tissues show
their own specific pattern of the amino-acid absorption but such normal-cell functions
may be impaired by amino acids abnormally excreted from neighboring tumor cells
(Douvlis, 1999).

4.4 Metabolic changes in response to inflammation


Tumor-associated persistent inflammatory responses are regarded as one of the causative
factors for changes in amino-acid metabolism. In addition, inflammation-mediated
proliferation of immune competent cells and synthesis of various inflammatory proteins
including cytokines and chemokines are also involved (Fox et al, 2005). In almost all of the
solid tumors, a variety of inflammatory responses are shown to be evoked in the tumor-
surrounding tissues (Mantovani et al, 2008). The tumor-associated inflammation is
characterized by a mixture of the anti-tumor inflammatory response, which directs tumor-
cell killing, and the tumor-induced inflammation, which stimulates tumor-cell proliferation
and promotes neovascularization (Schetter et al, 2010). Therefore, such tumor-associated
metabolic changes can be substantially different from those observed in other inflammatory
processes seen in pneumonia and colitis. In addition, these tumor-associated inflammations
can induce immunocompetent-cell proliferation and antibody production both in tumorous
tissues, regional lymph nodes, and the bone marrows, leading to further changes in
metabolism (Youn & Gabrilovich, 2010).

4.5 Metabolic changes in various remote normal organs


With an increase in amino-acid demands closely associated with elevated synthesis of
nucleotides and proteins in tumor cells, the amounts of amino acids recruited from other
tissues and organs are increased by means of enhancing whole body protein turnover,
elevating hepatic nonessential amino-acid biosynthesis, and stimulating proteolysis in
skeletal muscles accompanied by reduced protein synthesis (Rossi Fanelli et al, 1995). It is
well known that in liver and skeletal muscles, tumor-induced negative nitrogen balance can
promote intracellular production and extracellular release of glutamine (Medina et al, 1992).
In addition, it was reported that in a chronic inflammatory process, the amino-acid
metabolism could be influenced even in many remote organs: For instance, absorption of
blood cysteine and methionine were both elevated immediately after glutathione synthesis
was increased in the liver (Mercier et al, 2002).
306 Metabolomics

5. Future expectations
Although the applications of “AminoIndex technology” are still limited, the foundations for
their use for diagnostic purposes are in progress as described above. Studies with clinical
data indicate that even with individual variability, the “AminoIndex technology” can be
used to separate certain disease and physiological states. We believe that the amino acids are
a convenient metabolomic subset to use as a model for the development of metabolomics
based diagnostics, and that in the near future, other metabolites could be added to the
current analytical platform as practical issues such as stability are solved. At the same time,
the universality of the findings must be examined and it should be studied whether the data
set we have obtained for the Japanese population is applicable to other populations. We
believe that there is a great potential to use metabolome based markers in preliminary
diagnostic screening for multiple diseases in which a single measurement of a metabolomic
subset can lead to multiple diagnoses. One further advantage of the focused metabolomics
multiple metabolite marker approach is that since the biomarkers are generated from a
combination of already measured markers, new markers can be generated against any
measured target parameter. This means that if a focused metabolomic subset data is
obtained at the beginning of a treatment or an experiment, the generation of predictive
markers can be attempted with the outcome of the treatment or experiment as the target
parameter. We believe this would be of great use in tailor-made medicine and nutrition, as it
may be possible to discriminate populations for which certain pharmaceutical or nutritional
interventions would be useful or not.

6. References
Alpert, A. J. (1990) Hydrophilic-interaction chromatography for the separation of peptides,
nucleic acids and other polar compounds. J Chromatogr 499 pp.177-196
Aranibar, N., Borys, M., Mackin, N. A., Ly, V., Abu-Absi, N., Abu-Absi, S., Niemitz, M.,
Schilling, B., Li, Z. J., Brock, B., Russell, R. J., 2nd, Tymiak, A.&Reily, M. D. (2011)
NMR-based metabolomics of mammalian cell and tissue cultures. J Biomol NMR
49:(3-4) pp.195-206
Aspinall, R. J.&Pockros, P. J. (2004) The management of side-effects during therapy for
hepatitis C. Aliment Pharmacol Ther 20:(9) pp.917-29
Bapat, S. A., Krishnan, A., Ghanate, A. D., Kusumbe, A. P.&Kalra, R. S. (2010) Gene
expression: protein interaction systems network modeling identifies
transformation-associated molecules and pathways in ovarian cancer. Cancer Res
70:(12) pp.4809-19
Barr, J., Vazquez-Chantada, M., Alonso, C., Perez-Cormenzana, M., Mayo, R., Galan, A.,
Caballeria, J., Martin-Duce, A., Tran, A., Wagner, C., Luka, Z., Lu, S. C., Castro, A.,
Le Marchand-Brustel, Y., Martinez-Chantar, M. L., Veyrie, N., Clement, K.,
Tordjman, J., Gual, P.&Mato, J. M. (2010) Liquid chromatography-mass
spectrometry-based parallel metabolic profiling of human and mouse model serum
reveals putative biomarkers associated with the progression of nonalcoholic fatty
liver disease. J Proteome Res 9:(9) pp.4501-12
Bollard, M. E., Holmes, E., Lindon, J. C., Mitchell, S. C., Branstetter, D., Zhang,
W.&Nicholson, J. K. (2001) Investigations into biochemical changes due to diurnal
Clinical Implementation of Metabolomics 307

variation and estrus cycle in female rats using high-resolution (1)H NMR
spectroscopy of urine and pattern recognition. Anal Biochem 295:(2) pp.194-202
Bou Khalil, M., Hou, W., Zhou, H., Elisma, F., Swayne, L. A., Blanchard, A. P., Yao, Z.,
Bennett, S. A.&Figeys, D. (2010) Lipidomics era: accomplishments and challenges.
Mass Spectrom Rev 29:(6) pp.877-929
Bucci, M. (2011) Lipidomics: A viral egress. Nat Chem Biol 7:(9) pp.577
Caesar, R., Manieri, M., Kelder, T., Boekschoten, M., Evelo, C., Muller, M., Kooistra, T., Cinti,
S., Kleemann, R.&Drevon, C. A. (2010) A combined transcriptomics and lipidomics
analysis of subcutaneous, epididymal and mesenteric adipose tissue reveals
marked functional differences. PLoS One 5:(7) pp.e11525
Caruana, R., Niculescu-Mizil, A. (2006) An Empirical Comparison of Supervised Learning
Algorithms. In ICML2006 pp 161-168
Cascino, A., Muscaritoli, M., Cangiano, C., Conversano, L., Laviano, A., Ariemma, S.,
Meguid, M. M.&Rossi Fanelli, F. (1995) Plasma amino acid imbalance in patients
with lung and breast cancer. Anticancer Res 15:(2) pp.507-10
Castera, L., Vergniol, J., Foucher, J., Le Bail, B., Chanteloup, E., Haaser, M., Darriet, M.,
Couzigou, P.&De Ledinghen, V. (2005) Prospective comparison of transient
elastography, Fibrotest, APRI, and liver biopsy for the assessment of fibrosis in
chronic hepatitis C. Gastroenterology 128:(2) pp.343-50
Cynober, L. A. (2004) Metabolic and therapeutic aspects of amino acids in clinical nutrition, 2nd
edn, CRC Press, 0-8493-1382-1, Roca Raton
Dang, C. V. (2010) Rethinking the Warburg effect with Myc micromanaging glutamine
metabolism. Cancer Res 70:(3) pp.859-62
DeFeo, E. M., Wu, C. L., McDougal, W. S.&Cheng, L. L. (2011) A decade in prostate cancer:
from NMR to metabolomics. Nat Rev Urol 8:(6) pp.301-11
Dennis, E. A. (2009) Lipidomics joins the omics evolution. Proc Natl Acad Sci U S A 106:(7)
pp.2089-90
Douvlis, Z. (1999) Interference of amino acid patterns and tissue-specific amino acids
absorption dominance under the influence of tumor cell protein degradation
toxins. Med Hypotheses 53:(5) pp.450-7
Duda, R. O., Hart, P. E., Stork., D. G. (2001) Pattern Classification, 2nd edn, Wiley-
Interscience, 0-471-05669-3, New York
Dunn, W. B., Broadhurst, D. I., Atherton, H. J., Goodacre, R.&Griffin, J. L. (2011) Systems
level studies of mammalian metabolomes: the roles of mass spectrometry and
nuclear magnetic resonance spectroscopy. Chem Soc Rev 40:(1) pp.387-426
Fiehn, O. (2002) Metabolomics--the link between genotypes and phenotypes. Plant Mol Biol
48:(1-2) pp.155-71
Filho, J. C., Bergstrom, J., Stehle, P.&Furst, P. (1997) Simultaneous measurements of free
amino acid patterns of plasma, muscle and erythrocytes in healthy human subjects.
Clin Nutr 16:(6) pp.299-305
Fischer, J. E., Funovics, J. M., Aguirre, A., James, J. H., Keane, J. M., Wesdorp, R. I.,
Yoshimura, N.&Westman, T. (1975) The role of plasma amino acids in hepatic
encephalopathy. Surgery 78:(3) pp.276-90
308 Metabolomics

Fischer, J. E., Rosen, H. M., Ebeid, A. M., James, J. H., Keane, J. M.&Soeters, P. B. (1976) The
effect of normalization of plasma amino acids on hepatic encephalopathy in man.
Surgery 80:(1) pp.77-91
Fonteh, A. N., Harrington, R. J., Huhmer, A. F., Biringer, R. G., Riggins, J. N.&Harrington,
M. G. (2006) Identification of disease markers in human cerebrospinal fluid using
lipidomic and proteomic methods. Dis Markers 22:(1-2) pp.39-64
Forslund, A. H., Hambraeus, L., van Beurden, H., Holmback, U., El-Khoury, A. E., Hjorth,
G., Olsson, R., Stridsberg, M., Wide, L., Akerfeldt, T., Regan, M.&Young, V. R.
(2000) Inverse relationship between protein intake and plasma free amino acids in
healthy men at physical exercise. Am J Physiol Endocrinol Metab 278:(5) pp.E857-67
Fox, C. J., Hammerman, P. S.&Thompson, C. B. (2005) Fuel feeds function: energy
metabolism and the T-cell response. Nat Rev Immunol 5:(11) pp.844-52
Fried, M. W. (2002) Side effects of therapy of hepatitis C and their management. Hepatology
36:(5 Suppl 1) pp.S237-44
German, J. B., Gillies, L. A., Smilowitz, J. T., Zivkovic, A. M.&Watkins, S. M. (2007)
Lipidomics and lipid profiling in metabolomics. Curr Opin Lipidol 18:(1) pp.66-71
Grimplet, J., Cramer, G. R., Dickerson, J. A., Mathiason, K., Van Hemert, J.&Fennell, A. Y.
(2009) VitisNet: "Omics" integration through grapevine molecular networks. PLoS
One 4:(12) pp.e8365
Gruning, N. M., Lehrach, H.&Ralser, M. (2010) Regulatory crosstalk of the metabolic
network. Trends Biochem Sci 35:(4) pp.220-7
Gu, H., Pan, Z., Xi, B., Asiago, V., Musselman, B.&Raftery, D. (2011) Principal component
directed partial least squares analysis for combining nuclear magnetic resonance
and mass spectrometry data in metabolomics: application to the detection of breast
cancer. Anal Chim Acta 686:(1-2) pp.57-63
Han, X., Rozen, S., Boyle, S. H., Hellegers, C., Cheng, H., Burke, J. R., Welsh-Bohmer, K. A.,
Doraiswamy, P. M.&Kaddurah-Daouk, R. (2011) Metabolomics in early
Alzheimer's disease: identification of altered plasma sphingolipidome using
shotgun lipidomics. PLoS One 6:(7) pp.e21643
Hanley, J. A.&McNeil, B. J. (1982) The meaning and use of the area under a receiver
operating characteristic (ROC) curve. Radiology 143:(1) pp.29-36
Hilvo, M., Denkert, C., Lehtinen, L., Muller, B., Brockmoller, S., Seppanen-Laakso, T.,
Budczies, J., Bucher, E., Yetukuri, L., Castillo, S., Berg, E., Nygren, H., Sysi-Aho, M.,
Griffin, J. L., Fiehn, O., Loibl, S., Richter-Ehrenstein, C., Radke, C., Hyotylainen, T.,
Kallioniemi, O., Iljin, K.&Oresic, M. (2011) Novel theranostic opportunities offered
by characterization of altered membrane lipid metabolism in breast cancer
progression. Cancer Res 71:(9) pp.3236-45
Hulley, S. B., Cummings, R. B., Browner, W. S., Grady, D. G., Newman, T. B. (2006)
Designing Clinical Research: An Epidemiologic Approach 3rd edn, Lippincott Williams
& Willkins, Inc., Philadelphia
Hunter, P. (2009) Reading the metabolic fine print. The application of metabolomics to
diagnostics, drug research and nutrition might be integral to improved health and
personalized medicine. EMBO Rep 10:(1) pp.20-3
Clinical Implementation of Metabolomics 309

Imbert-Bismut, F., Ratziu, V., Pieroni, L., Charlotte, F., Benhamou, Y.&Poynard, T. (2001)
Biochemical markers of liver fibrosis in patients with hepatitis C virus infection: a
prospective study. Lancet 357:(9262) pp.1069-75
Ishii, N., Nakahigashi, K., Baba, T., Robert, M., Soga, T., Kanai, A., Hirasawa, T., Naba, M.,
Hirai, K., Hoque, A., Ho, P. Y., Kakazu, Y., Sugawara, K., Igarashi, S., Harada, S.,
Masuda, T., Sugiyama, N., Togashi, T., Hasegawa, M., Takai, Y., Yugi, K., Arakawa,
K., Iwata, N., Toya, Y., Nakayama, Y., Nishioka, T., Shimizu, K., Mori, H.&Tomita,
M. (2007) Multiple high-throughput analyses monitor the response of E. coli to
perturbations. Science 316:(5824) pp.593-7
Jenkins, H., Hardy, N., Beckmann, M., Draper, J., Smith, A. R., Taylor, J., Fiehn, O.,
Goodacre, R., Bino, R. J., Hall, R., Kopka, J., Lane, G. A., Lange, B. M., Liu, J. R.,
Mendes, P., Nikolau, B. J., Oliver, S. G., Paton, N. W., Rhee, S., Roessner-Tunali, U.,
Saito, K., Smedsgaard, J., Sumner, L. W., Wang, T., Walsh, S., Wurtele, E. S.&Kell,
D. B. (2004) A proposed framework for the description of plant metabolomics
experiments and their results. Nat Biotechnol 22:(12) pp.1601-6
Kaddurah-Daouk, R., Kristal, B. S.&Weinshilboum, R. M. (2008) Metabolomics: a global
biochemical approach to drug response and disease. Annu Rev Pharmacol Toxicol 48
pp.653-83
Kano, T., Nagaki, M., Takahashi, T., Ohnishi, H., Saitoh, K., Kimura, K.&Muto, Y. (1991)
Plasma free amino acid pattern in chronic hepatitis as a sensitive and prognostic
index. Gastroenterol Jpn 26:(3) pp.344-9
Kell, D. B. (2002) Metabolomics and machine learning: explanatory analysis of complex
metabolome data using genetic programming to produce simple, robust rules. Mol
Biol Rep 29:(1-2) pp.237-41
Kell, D. B. (2006) Systems biology, metabolic modelling and metabolomics in drug discovery
and development. Drug Discov Today 11:(23-24) pp.1085-92
Kim, Y., Koo, I., Jung, B. H., Chung, B. C.&Lee, D. (2010) Multivariate classification of urine
metabolome profiles for breast cancer diagnosis. BMC Bioinformatics 11 Suppl 2
pp.S4
Kimura, T., Noguchi, Y., Shikata, N.&Takahashi, M. (2009) Plasma amino acid analysis for
diagnosis and amino acid-based metabolic networks. Curr Opin Clin Nutr Metab
Care 12:(1) pp.49-53
Kirouac, D. C., Ito, C., Csaszar, E., Roch, A., Yu, M., Sykes, E. A., Bader, G. D.&Zandstra, P.
W. (2010) Dynamic interaction networks in a hierarchically organized tissue. Mol
Syst Biol 6 pp.417
Lai, H. S., Lee, J. C., Lee, P. H., Wang, S. T.&Chen, W. J. (2005) Plasma free amino acid
profile in cancer patients. Semin Cancer Biol 15:(4) pp.267-76
Lee, J. C., Chen, M. J., Chang, C. H., Tiai, Y. F., Lin, P. W., Lai, H. S.&Wang, S. T. (2003)
Plasma amino acid levels in patients with colorectal cancers and liver cirrhosis with
hepatocellular carcinoma. Hepatogastroenterology 50:(53) pp.1269-73
Lin, L., Huang, Z., Gao, Y., Yan, X., Xing, J.&Hang, W. (2011a) LC-MS based serum
metabonomic analysis for renal cell carcinoma diagnosis, staging, and biomarker
discovery. J Proteome Res 10:(3) pp.1396-405
310 Metabolomics

Lin, Z. H., Xin, Y. N., Dong, Q. J., Wang, Q., Jiang, X. J., Zhan, S. H., Sun, Y.&Xuan, S. Y.
(2011b) Performance of the aspartate aminotransferase-to-platelet ratio index for
the staging of hepatitis C-related fibrosis: an updated meta-analysis. Hepatology
53:(3) pp.726-36
Lu, X., Zhao, X., Bai, C., Zhao, C., Lu, G.&Xu, G. (2008) LC-MS-based metabonomics
analysis. J Chromatogr B Analyt Technol Biomed Life Sci 866:(1-2) pp.64-76
Maeda, J., Higashiyama, M., Imaizumi, A., Nakayama, T., Yamamoto, H., Daimon, T.,
Yamakado, M., Imamura, F.&Kodama, K. (2010) Possibility of multivariate function
composed of plasma amino acid profiles as a novel screening index for non-small
cell lung cancer: a case control study. BMC Cancer 10:(1) pp.690
Mantovani, A., Allavena, P., Sica, A.&Balkwill, F. (2008) Cancer-related inflammation.
Nature 454:(7203) pp.436-44
Matthew, E. M., Hart, L. S., Astrinidis, A., Navaraj, A., Dolloff, N. G., Dicker, D. T., Henske,
E. P.&El-Deiry, W. S. (2009) The p53 target Plk2 interacts with TSC proteins
impacting mTOR signaling, tumor growth and chemosensitivity under hypoxic
conditions. Cell Cycle 8:(24) pp.4168-75
Medina, M. A., Marquez, J.&Nunez de Castro, I. (1992) Interchange of amino acids between
tumor and host. Biochem Med Metab Biol 48:(1) pp.1-7
Mercier, S., Breuille, D., Mosoni, L., Obled, C.&Patureau Mirand, P. (2002) Chronic
inflammation alters protein metabolism in several organs of adult rats. J Nutr
132:(7) pp.1921-8
Metavir. (1994) Intraobserver and interobserver variations in liver biopsy interpretation in
patients with chronic hepatitis C. The French METAVIR Cooperative Study Group.
Hepatology 20:(1 Pt 1) pp.15-20
Min, H. K., Lim, S., Chung, B. C.&Moon, M. H. (2011) Shotgun lipidomics for candidate
biomarkers of urinary phospholipids in prostate cancer. Anal Bioanal Chem 399:(2)
pp.823-30
Momin, A. A., Park, H., Portz, B. J., Haynes, C. A., Shaner, R. L., Kelly, S. L., Jordan, I.
K.&Merrill, A. H., Jr. (2011) A method for visualization of "omic" datasets for
sphingolipid metabolism to predict potentially interesting differences. J Lipid Res
52:(6) pp.1073-83
Montoliu, I., Martin, F. P., Collino, S., Rezzi, S.&Kochhar, S. (2009) Multivariate modeling
strategy for intercompartmental analysis of tissue and plasma 1H NMR
spectrotypes. J Proteome Res 8:(5) pp.2397-406
Moore, S., Spackman, D. H.&Stein, W. H. (1958) Automatic recording apparatus for use in
the chromatography of amino acids. Fed Proc 17:(4) pp.1107-15
Naini, A. B., Dickerson, J. W.&Brown, M. M. (1988) Preoperative and postoperative levels of
plasma protein and amino acid in esophageal and lung cancer patients. Cancer
62:(2) pp.355-60
Noguchi, Y., Shikata, N., Furuhata, Y., Kimura, T.&Takahashi, M. (2008) Characterization of
dietary protein-dependent amino acid metabolism by linking free amino acids with
transcriptional profiles through analysis of correlation. Physiol Genomics 34:(3)
pp.315-26
Clinical Implementation of Metabolomics 311

Norton, J. A., Gorschboth, C. M., Wesley, R. A., Burt, M. E.&Brennan, M. F. (1985) Fasting
plasma amino acid levels in cancer patients. Cancer 56:(5) pp.1181-6
Okamoto, N., Miyagi, Y., Chiba, A., Akaike, M., Shiozawa, M., Imaizumi, A.,
Yamamoto, H., Ando, T., Yamakado, M.&Tochikubo, O. (2009) Diagnostic
modeling with differences in plasma amino acid profiles between non-cachectic
colorectal/breast cancer patients and healthy individuals. Int J Med Med Sci
1:(1) pp.1-8
Piraud, M., Vianey-Saban, C., Petritis, K., Elfakir, C., Steghens, J. P., Morla, A.&Bouchu, D.
(2003) ESI-MS/MS analysis of underivatised amino acids: a new tool for the
diagnosis of inherited disorders of amino acid metabolism. Fragmentation study of
79 molecules of biological interest in positive and negative ionisation mode. Rapid
Commun Mass Spectrom 17:(12) pp.1297-311
Plumb, R. S., Stumpf, C. L., Granger, J. H., Castro-Perez, J., Haselden, J. N.&Dear, G. J. (2003)
Use of liquid chromatography/time-of-flight mass spectrometry and multivariate
statistical analysis shows promise for the detection of drug metabolites in biological
fluids. Rapid Commun Mass Spectrom 17:(23) pp.2632-8
Poynard, T., Yuen, M. F., Ratziu, V.&Lai, C. L. (2003) Viral hepatitis C. Lancet 362:(9401)
pp.2095-100
Proenza, A. M., Oliver, J., Palou, A.&Roca, P. (2003) Breast and lung cancer are
associated with a decrease in blood cell amino acid content. J Nutr Biochem
14:(3) pp.133-8
Puri, P., Wiest, M. M., Cheung, O., Mirshahi, F., Sargeant, C., Min, H. K., Contos, M. J.,
Sterling, R. K., Fuchs, M., Zhou, H., Watkins, S. M.&Sanyal, A. J. (2009) The
plasma lipidomic signature of nonalcoholic steatohepatitis. Hepatology 50:(6)
pp.1827-38
Qu, Y., Slocum, R. H., Fu, J., Rasmussen, W. E., Rector, H. D., Miller, J. B.&Coldwell, J. G.
(2001) Quantitative amino acid analysis using a Beckman system gold HPLC
126AA analyzer. Clin Chim Acta 312:(1-2) pp.153-62
Quinones, M. P.&Kaddurah-Daouk, R. (2009) Metabolomics tools for identifying biomarkers
for neuropsychiatric diseases. Neurobiol Dis 35:(2) pp.165-76
Rhee, E. P., Cheng, S., Larson, M. G., Walford, G. A., Lewis, G. D., McCabe, E., Yang, E.,
Farrell, L., Fox, C. S., O'Donnell, C. J., Carr, S. A., Vasan, R. S., Florez, J. C., Clish, C.
B., Wang, T. J.&Gerszten, R. E. (2011) Lipid profiling identifies a triacylglycerol
signature of insulin resistance and improves diabetes prediction in humans. J Clin
Invest 121:(4) pp.1402-11
Righi, V., Durante, C., Cocchi, M., Calabrese, C., Di Febo, G., Lecce, F., Pisi, A., Tugnoli, V.,
Mucci, A.&Schenetti, L. (2009) Discrimination of healthy and neoplastic human
colon tissues by ex vivo HR-MAS NMR spectroscopy and chemometric analyses. J
Proteome Res 8:(4) pp.1859-69
Rossi Fanelli, F., Cangiano, C., Muscaritoli, M., Conversano, L., Torelli, G. F.&Cascino, A.
(1995) Tumor-induced changes in host metabolism: a possible marker of neoplastic
disease. Nutrition 11:(5 Suppl) pp.595-600
312 Metabolomics

Roux, A., Lison, D., Junot, C.&Heilier, J. F. (2011) Applications of liquid chromatography
coupled to mass spectrometry-based metabolomics in clinical chemistry and
toxicology: A review. Clin Biochem 44:(1) pp.119-35
Schetter, A. J., Heegaard, N. H.&Harris, C. C. (2010) Inflammation and cancer:
interweaving microRNA, free radical, cytokine and p53 pathways.
Carcinogenesis 31:(1) pp.37-49
Serkova, N. J., Standiford, T. J.&Stringer, K. A. (2011) The Emerging Field of Quantitative
Blood Metabolomics for Biomarker Discovery in Critical Illnesses. Am J Respir Crit
Care Med
Shiffman, M. L. (2004) Management of patients with chronic hepatitis C virus infection and
previous nonresponse. Rev Gastroenterol Disord 4 Suppl 1 pp.S22-30
Shimbo, K., Kubo, S., Harada, Y., Oonuki, T., Yokokura, T., Yoshida, H., Amao, M.,
Nakamura, M., Kageyama, N., Yamazaki, J., Ozawa, S. I., Hirayama, K., Ando, T.,
Miura, J.&Miyano, H. (2009a) Automated precolumn derivatization system for
analyzing physiological amino acids by liquid chromatography/mass
spectrometry. Biomed Chromatogr 24:(7) pp.683-91
Shimbo, K., Oonuki, T., Yahashi, A., Hirayama, K.&Miyano, H. (2009b) Precolumn
derivatization reagents for high-speed analysis of amines and amino acids in
biological fluid using liquid chromatography/electrospray ionization tandem mass
spectrometry. Rapid Commun Mass Spectrom 23:(10) pp.1483-92
Shimbo, K., Yahashi, A., Hirayama, K., Nakazawa, M.&Miyano, H. (2009c)
Multifunctional and highly sensitive precolumn reagents for amino acids in
liquid chromatography/tandem mass spectrometry. Anal Chem 81:(13) pp.5172-
9
Soga, T. (2007) Capillary electrophoresis-mass spectrometry for metabolomics. Methods Mol
Biol 358 pp.129-37
Spagou, K., Wilson, I. D., Masson, P., Theodoridis, G., Raikos, N., Coen, M., Holmes, E.,
Lindon, J. C., Plumb, R. S., Nicholson, J. K.&Want, E. J. (2011) HILIC-UPLC-MS for
exploratory urinary metabolic profiling in toxicological studies. Anal Chem 83:(1)
pp.382-90
Spratlin, J. L., Serkova, N. J.&Eckhardt, S. G. (2009) Clinical applications of metabolomics in
oncology: a review. Clin Cancer Res 15:(2) pp.431-40
Steuer, R. (2006) Review: on the analysis and interpretation of correlations in metabolomic
data. Brief Bioinform 7:(2) pp.151-8
Sugimoto, M., Wong, D. T., Hirayama, A., Soga, T.&Tomita, M. (2010) Capillary
electrophoresis mass spectrometry-based saliva metabolomics identified oral,
breast and pancreatic cancer-specific profiles. Metabolomics 6:(1) pp.78-95
Taylor, N. S., Weber, R. J., White, T. A.&Viant, M. R. (2010) Discriminating between different
acute chemical toxicities via changes in the daphnid metabolome. Toxicol Sci 118:(1)
pp.307-17
Taylor, R.&Singhal, M. (2009) Biological Network Inference and analysis using SEBINI and
CABIN. Methods Mol Biol 541 pp.551-76
Thysell, E., Surowiec, I., Hornberg, E., Crnalic, S., Widmark, A., Johansson, A. I., Stattin, P.,
Bergh, A., Moritz, T., Antti, H.&Wikstrom, P. (2010) Metabolomic characterization
Clinical Implementation of Metabolomics 313

of human prostate cancer bone metastases reveals increased levels of cholesterol.


PLoS One 5:(12) pp.e14175
Tsukamoto, Y., Santa, T., Yoshida, H., Miyano, H., Fukushima, T., Hirayama, K., Imai,
K.&Funatsu, T. (2006) Synthesis of the isotope-labeled derivatization reagent for
carboxylic acids, 7-(N,N-dimethylaminosulfonyl)-4-(aminoethyl)piperazino-2,1,3-
benzoxadiazol e (d6) [DBD-PZ-NH2 (D)], and its application to the quantification
and the determination of relative amount of fatty acids in rat plasma samples by
high-performance liquid chromatography/mass spectrometry. Biomed Chromatogr
20:(4) pp.358-64
Valdes-Gonzalez, T., Goto-Inoue, N., Hirano, W., Ishiyama, H., Hayasaka, T., Setou,
M.&Taki, T. (2011) New approach for glyco- and lipidomics--molecular scanning of
human brain gangliosides by TLC-Blot and MALDI-QIT-TOF MS. J Neurochem
116:(5) pp.678-83
van der Greef, J., Hankemeier, T.&McBurney, R. N. (2006) Metabolomics-based systems
biology and personalized medicine: moving towards n = 1 clinical trials?
Pharmacogenomics 7:(7) pp.1087-94
Vissers, Y. L., Dejong, C. H., Luiking, Y. C., Fearon, K. C., von Meyenfeldt, M. F.&Deutz, N.
E. (2005) Plasma arginine concentrations are reduced in cancer patients: evidence
for arginine deficiency? Am J Clin Nutr 81:(5) pp.1142-6
Wheelock, C. E., Wheelock, A. M., Kawashima, S., Diez, D., Kanehisa, M., van Erk, M.,
Kleemann, R., Haeggstrom, J. Z.&Goto, S. (2009) Systems biology approaches and
pathway tools for investigating cardiovascular disease. Mol Biosyst 5:(6) pp.588-
602
Wise, D. R.&Thompson, C. B. (2010) Glutamine addiction: a new therapeutic target in
cancer. Trends Biochem Sci 35:(8) pp.427-33
Yang, W. C., Mirzaei, H., Liu, X.&Regnier, F. E. (2006) Enhancement of amino acid detection
and quantification by electrospray ionization mass spectrometry. Anal Chem 78:(13)
pp.4702-8
Yoshida, H., Mizukoshi, T., Hirayama, K.&Miyano, H. (2007) Comprehensive analytical
method for the determination of hydrophilic metabolites by high-performance
liquid chromatography and mass spectrometry. J Agric Food Chem 55:(3) pp.551-
60
Youn, J. I.&Gabrilovich, D. I. (2010) The biology of myeloid-derived suppressor cells: the
blessing and the curse of morphological and functional heterogeneity. Eur J
Immunol 40:(11) pp.2969-75
Yuan, K., Kong, H., Guan, Y., Yang, J.&Xu, G. (2007) A GC-based metabonomics
investigation of type 2 diabetes by organic acids metabolic profile. J Chromatogr B
Analyt Technol Biomed Life Sci 850:(1-2) pp.236-40
Zeisel, S. H. (2007) Nutrigenomics and metabolomics will change clinical nutrition and
public health practice: insights from studies on dietary requirements for choline.
Am J Clin Nutr 86:(3) pp.542-8
Zhang, G. F., Sadhukhan, S., Tochtrop, G. P.&Brunengraber, H. (2011) Metabolomics,
pathway regulation, and pathway discovery. J Biol Chem 286:(27) pp.23631-5
314 Metabolomics

Zhang, P. C.&Pang, C. P. (1992) Plasma amino acid patterns in cancer. Clin Chem 38:(6)
pp.1198-9
Zhang, Q., Takahashi, M., Noguchi, Y., Sugimoto, T., Kimura, T., Okumura, A., Ishikawa,
T.&Kakumu, S. (2006) Plasma amino acid profiles applied for diagnosis of
advanced liver fibrosis in patients with chronic hepatitis C infection. Hepatol Res
34:(3) pp.170-7
Part 6

Improving Analytics
13

Improvement in the Number of Analytic


Features Detected by Non-Targeted
Metabolomic Analysis: Influence of
the Chromatographic System and
the Ionization Technique
R. Pandher1, E. Naegele2, S.M. Fischer3 and F.I Raynaud1
1The Institute of Cancer Research, Pharmacokinetics and Metabolomics,
2Agilent Technologies Research and Development, Waldbronn,
3Agilent Technologies, Metabolomics Laboratory, Santa Clara, CA,
1UK
2Germany
3USA

1. Introduction
The development and optimisation of genomic, transcriptomic and proteomic technologies
have significantly contributed to the assessment of biological systems and increased our
understanding of gene function and regulation (Kitano 2002, Brown et al., 1999, Pandey and
Mann 2000). In addition, metabolic fingerprinting or metabolomics complement these
approaches by measuring low molecular weight chemicals in biological samples (Nicholson
and Lindon, 2008). The elucidation of the links between genetic regulation, kinetic activities
of enzymes and metabolic reactions is key to understanding homeostatic regulation of living
organisms and the effects of food, diurnal variations disease and drugs (Nicholson et al.,
2003, van der Greef et al., 2003, Plumb et al., 2003). Mapping of these various interactions is
likely to result in applications in disciplines such as agriculture and medicine (Lee et al.,
2007, Borodina and Nielson 2007, Wishart 200, Ducruix et al., 2006). Several analytic tools
have been applied to profile the metabolome.
LC-MS studies are a more recent introduction to the field of metabolomics compared with the
more established techniques of GC-MS and NMR. LC-MS can be used for the analysis of
metabolites with a wide range of molecular weights than those detectable by GC-MS including
polar and non-volatile compounds. With LC-MS, many different chromatographic phases and
thus separation techniques are available when compared with GC-MS (Dunn, 2008).
Targeted metabolomic studies allow the identification and quantification of defined sets of
metabolites and are performed using triple Quadrupole mass spectrometers which provide
sensitivity and selectivity. Non-targeted global metabolomic studies are carried out on
instruments with good mass accuracy such as time of flight and orbitrap mass analysers. In
318 Metabolomics

metabolomic profiling, comparison of biological samples collected under different


conditions is performed by multivariate statistical analysis in order to to identify significant
differences between the groups. In metabolomics a “feature” is a molecular ion (m/z)
coupled to a retention time (RT) that is generated following data processing, where feature
finding is performed in conjunction with noise reduction and alignment of data. A drift in
mass accuracy or retention time will affect the experimental results by creating additional
numbers of novel features. An increase in variability of peak area or height may mask
differences between experimental groups. The number of features, and their intensities in a
number of replicate analysis of a given sample can define the robustness of an analytical run
prior to complex statistical analysis of the data. . The acceptance criteria for reproducibility
and repeatability differ between laboratories. In the studies presented in this chapter a
coefficient of variation (CV) threshold of 25% was set which is in line with similar
metabolomic studies published in the literature (Crews et al., 2009; Lai et al., 2010).
Plasma represents an important biofluid and global metabolite profiles have been derived
from a variety of LCMS methods (Sabatine et al., 2005, Want et al., 2006, Bruce et al., 2008
and Zelena et al., 2009). We have previously shown that 2 different QTOF instruments
produced the same number of reproducible features from tissue culture media extracts
(Pandher et al., 2009). The goal of this study as to develop an efficient methodology using
the Agilent LC Infinity system and Jet Stream Technology for metabolomic reverse phase
LC-MS approaches. Here we describe the number of features obtained in human plasma
extracts with a conventional rapid resolution chromatographic system and a QTOF mass
spectrometer equipped with an electrospray ionization source in positive ionization mode.
The improvement in the number of features and reproducibility following chromatographic
separation with the Agilent 1290 Infinity LC system at various flow rates is also presented
together with the peak capacity of a selected number of analytes. In addition, the impact of
further optimization of the analytical conditions (temperature, flow rate) with the Jet Stream
ionization technology on the number and reproducibility of the ions detected is presented.

2. Materials and method


Water (LC-MS grade), acetonitrile (LC-MS grade) and formic acid (Aristar grade) were all
purchased from Fisher Scientific (Loughborough, UK). Leucine enkephalin was purchased
from Sigma (Poole, UK). The external standards creatine (CAS no: 57-00-1), carnitine (CAS
no: 541-14-0), colchicine (CAS no: 64-86-8), hydrocortisone (CAS no: 50-23-7), phenylalanine
(CAS no: 673-06-3), tryptophan (CAS: 73-22-3) and hippuric acid (CAS no: 495-69-2) were
purchased from Sigma (Poole, UK). Standard stock solutions of 1 mM were prepared in
water or DMSO as appropriate and in human plasma. Human plasma was collected in
heparinised tubes from healthy donors and centrifuged at 1500 x g for 15 minutes at 4°C.
Plasma was then stored at -80°C until analysis.

3. Sample extraction
Plasma samples (200 μl) were extracted with 4 volumes of acetonitrile, using a 96 well
protein precipitation plate (Whatman, Maidstone, UK). The plate was vortexed for 1 min
before a vacuum was applied. The filtered samples were collected in a 96 deep well plate
and plasma extracts were pooled and aliquoted out for further analysis.
Improvement in the Number of Analytic Features Detected by Non-Targeted
Metabolomic Analysis: Influence of the Chromatographic System and the Ionization Technique 319

3.1 Sample analysis


For optimization of the analytical conditions, triplicate samples containing the spiked
analytical standards were injected. For reproducibility studies, 3 replicates of unspiked and
spiked plasma samples were analysed.

3.2 Liquid chromatographic separation


The HPLC systems used were the conventional Agilent 1200 and the Agilent 1290 Infinity
LC system. Most of the analytic separation was achieved on a Waters Acquity column HSS
T3 C18 (100 mm × 2.1 mm, I.D 1.8μm particles) and a 150 mm column was also tested.
Different chromatographic conditions were evaluated:
A mobile phase of 100% 0.1% formic acid was run isocratic for 0.5 minutes followed by a
linear gradient ending in 100% acetonitrile over 7.5 minutes or 5.5 minutes or 3.5 minutes,
followed by 100% acetonitrile over 2 minutes. After returning to the original conditions, the
system was left to equilibrate for 3 minutes prior to the next injection. Different flow rates
were evaluated (0.4 ml/min on the 1200 HPLC and 0.4, 0.6, 0.8 and 1 ml/min on the 1290
Infinity UHPLC). The same gradients were used on the 150 mm column. Columns were
previously equilibrated with the injection of 5 plasma extracts.

3.3 Mass spectrometric analysis


Mass spectrometry was performed in positive ionization mode on a QTOF (6530, Agilent).
Two different sources were evaluated: the classical electrospray ionization source and the Jet
Stream technology. With the ESI source, parameters were set with a capillary voltage of 4 kV
in positive ionisation mode. The fragmentor voltage was 140 V and skimmer was 65 V. The
gas temperature was 250°C, drying gas 10 l/min and nebulizer 40 psig. Nitrogen was used
as a drying gas. MS spectra were acquired in full scan analysis over an m/z range of 70-1000
using extended dynamic range and a scan rate of 1.4 spectra/second. To maintain mass
accuracy during the run time, a reference mass solution containing reference ions 121.0508
and 922.0097 was used.
With the Jet Stream technology; parameters were set with a capillary voltage of 4 kV in
positive ionisation mode. The fragmentor voltage was 140 V and skimmer was 65 V. The gas
temperature was 250°C, drying gas 6 l/min and nebulizer 60 psig. Nitrogen was used as a
drying gas. The sheath gas temperature was tested and optimized from 200°C to 400°C by
increments of 50°C and the sheath gas flow rate was 11l/min. Total ion spectra were
acquired in full scan analysis over an m/z range of 70-1000 using extended dynamic range
2GHz and an acquisition rate of 2Hz.
Sampling rates of 2, 4 and 6Hz were tested with the Jet Stream technology at 0.6 ml/min
and 0.8 ml/min with the sheath gas at 200°C in order to evaluate the number of sampling
points collected across each peak and the reproducibility of the analysis.

4. Data processing
Sample features were extracted with the molecular feature extractor (MassHunter
Workstation Software (version B.01.03)). Data were processed using the following
320 Metabolomics

conditions: restrict retention time to 0.20 - 8.5 min, restrict m/z to 100-800, absolute height
threshold: 25000 or 2500, mass tolerance: 0.05, peaks with height: > 100 counts, isotope
grouping: peak spacing tolerance: 0.0025 m/z, plus 7.0 ppm, isotope model: common
organic model, mass filters: filter mass list: 20 ppm.
The list of features consisting of retention times (RT) and molecular masses was then
analysed using GeneSpring MS Analysis Platform (v1.2, Agilent Technologies, Inc., Santa
Clara, CA) where they were aligned and normalized.
Data were then imported into Excel spreadsheets and mean, SD and CV of all features was
calculated.

5. Results and dicussion


There are a number of experimental variables, related to chromatography or mass
spectrometry, that can impact the reproducibility of metabolomic profile data. This in turn
can compromise the validity of the data’s biological relevance and applicability. The
importance of several variables, including flow rate, column length, mass spectrometric
conditions were all evaluated in terms of the number of features found.
Following triplicate separation of human plasma on the conventional 1200 LC system at 0.4
ml/min with a 7.5 min gradient followed by 2 min isocratic on a 10 cm column with the ESI
source, 1324 total features were detected out of which 795 (60%) showed less than 25% CV
Table 1).

% of % change in features
Total no. No. of
System features with <25% relative
Method of features
pressure with to conventional
features <25% CV
<25% CV system

LC1200 0.4ml/min 450 1324 795 60 n/a

LC1290 0.4ml/min 270 1714 925 54 +16%

LC1290 0.6ml/min 390 2559 1149 45 +44%

LC1290 0.8ml/min 502 2263 1074 47 +35%

LC1290 1.0ml/min 605 1805 305 17 -38%

aChromatography performed on a reverse phase Waters Acquity T3 column with a 7.5min 0.1% formic

acid: acetonitrile gradient. Mass spectrometry analysis performed on 6530 QTOF in ESI mode. Data was
extracted using MassHunter Qualitative software package and GeneSpringMS and then exported to
Excel where statistics were performed.
Table 1. Effect of flow rate on the number and reproducibility of features present in technical
replicates of human plasma extracts conventional 1200 LC and novel 1290 Infinity LC in ESI
mode a (n=3).
Improvement in the Number of Analytic Features Detected by Non-Targeted
Metabolomic Analysis: Influence of the Chromatographic System and the Ionization Technique 321

Under similar conditions, the novel 1290 LC system generated 16% higher and
reproducible features compared with the conventional system without any change in
analytic conditions. Increasing the flow rate to 0.6 ml/min with the same gradient
increased the number of reproducible features to 1149 allowing a 44% improvement
when compared with the 1200 LC system. When the flow rate was increased from 0.4 to
0.6 ml/min many additional features were detected that were not previously observed
following separation by the 1200 system. Careful examination of the features showed
that they were mainly ions that had not previously eluted from the column at 0.4
ml/min. Further increase in flow rate showed that fewer reproducible features were
detected. The pressure in the system with the column installed was significantly lower in
the 1290 system with a back pressure of at 132 bar at 0.4 ml/min on the 1290 versus 450
bars on the 1200 respectively. At 1 ml/min, the pressure on the 1290 system was 605 bars
only. It is possible that the different composition of the pistons and their independent
operation together with the novel mixing technology used in the 1290 Infinity LC system
can explain the decreased pressures compared with the conventional 1200 (data not
shown)
The length of the gradient was then shortened to 5.5 minutes and 3.5 minutes
respectively but this resulted in a significant decrease in reproducibility (Table 2). In fact,
we noted that the isocratic segment of the gradient had to be extended in order to avoid
carry-over from previous samples which defeated the purpose of a shorter analytic run
(data not shown).

% of
Total no. No. of % change in features
features
Methodb of features with <25% relative to
with <25%
features <25% CV conventional system
CV

LC1200 0.4ml/min 7.5min gradient 1324 795 60 n/a

LC1290 0.6ml/min 7.5 min gradient 2559 1149 45 +44%

LC1290 0.6ml/min 5.5 min gradient 1989 1086 55 +37%

LC1290 0.6ml/min 3.5 min gradient 1889 1017 54 +28%

aChromatography performed on a reverse phase Waters Acquity T3 column with a 7.5min 0.1% formic

acid: acetonitrile gradient. Mass spectrometry analysis performed on 6530 QTOF in ESI mode. Data was
extracted using MassHunter Qualitative software package and GeneSpringMS and then exported to
Excel where statistics were performed.
Each gradient was preceded by 0.5min of 100% A (0.1% formic acid in water) and followed by 2min of 100% B
(0.1 formic acid in acetonitrile).
Table 2. The effect of gradient duration on the number and reproducibility of features
present in technical replicates of human plasma extracts using conventional 1200 and novel
1290 Infinity LC systems in ESI mode a (n=3).
322 Metabolomics

Following triplicate analysis of human plasma on the conventional 1200 LC system using
the 150 mm column with our previously described gradient, there was no significant
improvement in total or reproducible number of features when compared with the 100 mm
column regardless of the flow rate (data not shown). Our conclusion for the data from the
ESI source was that a flow rate of 0.6 ml/min was optimal with the 100 mm column with the
original 7.5 minute gradient.
We then proceeded to evaluate the effect of the Jet Stream technology on the number of
features detected and their repeatability. Incremental temperatures of 50°C of heated
nitrogen sheath gas; from 200°C to 400°C were applied and evaluated. At 0.6 ml/min
with a sheath gas of 200°C, both the total and reproducible features were more than
doubled when compared to the equivalent result with the ESI source. Overall, 50% of
features showed less 25% CV over triplicate analysis (Table 3). This represents a 173%
increase in reproducible features when compared with the conventional 1200 LC system
and the ESI source.

% of
Total no. No. of % change in features
features
Method of features with <25% relative to
with <25%
features <25% CV conventional system
CV

ESI LC1200 0.4ml/min 1324 795 60 n/a

JS LC1290 0.6ml/min 200°C 4357 2176 50 +173%

JS LC1290 0.8ml/min 200°C 4312 2512 58 +215%

JS LC1290 0.6ml/min 250°C 4396 2294 52 +189%

JS LC1290 0.8ml/min 250°C 4810 2708 56 +241%

JS LC1290 0.6ml/min 300°C 4463 2565 57 +223%

JS LC1290 0.8ml/min 300°C 5130 2869 56 +261%

JS LC1290 0.6ml/min 350°C 4693 2762 59 +247%

JS LC1290 0.8ml/min 350°C 5257 2994 57 +277%

JS LC1290 0.6ml/min 400°C 4919 2707 55 +241%

JS LC1290 0.8ml/min 400°C 5095 3310 65 +316%


aChromatography performed on a reverse phase Waters Acquity T3 column with a 7.5min 0.1% formic

acid: acetonitrile gradient. Mass spectrometry analysis performed on 6530 QTOF in ESI mode. Data was
extracted using MassHunter Qualitative software package and GeneSpringMS and then exported to
Excel where statistics were performed.
Table 3. The effect of Jet Stream (JS) sheath gas and flow rates on number and
reproducibility of features present in technical replicates of human plasma extracts the novel
1290 Infinity LC system coupled to a 6530 QTOF using Jet Stream technology a (n=3)
Improvement in the Number of Analytic Features Detected by Non-Targeted
Metabolomic Analysis: Influence of the Chromatographic System and the Ionization Technique 323

Fig. 1. Number of reproducible features against retention time. Number of reproducible


features generated following data processing using the conventional 1200 LC system with
ESI, the novel 1290 Infinity LC system with ESI and the novel LC system with Jet Stream
technology.

The increased number of features ionized by the Jet Stream technology when compared
with ESI is illustrated in Figure 1 which shows overlayed total ion chromatograms. In
contrast to our results with the ESI source, increasing the flow rate to 0.8 ml/min further
increased the number of reproducible features when compared with 0.6 ml/min (Table 3).
Increasing the temperature of the sheath gas by increments of 50°C at both 0.6 and 0.8
ml/min gradually increased the number of features. At 400°C, a striking 5095 features were
detected; 3310 of which showed less than 25% CV.
We were concerned that this significant increase in features with increase in temperature
could be the result of thermal degradation of ions. . To address this, a Venn diagram derived
from GeneSpring MS shows the ions present at at both temperatures, at 200°C or 400°C
only. (Figure 2).
The heat plot in Figure 3 demonstrates that all the 1491 ions present at 400° C were weak
whereas more than half of the 888 were stronger in intensity, suggesting that the increase
in temperature fragmented the ions and that thermal degradation occurred. Further
examination of the features at 200°C in Jet Stream and ESI at 0.6 ml/min, showed that
>2000 features were specific to Jet Stream alone. By analysing the most intense features
we found that they were not split features due to errors in the automatic processing
software. We were concerned that some of these features may be detectable in ESI at a
lower threshold. Therefore, we proceeded to lower the threshold to 2500 in both Jet
Stream and ESI and found that 10599 and 15424 total features and 2792 and 4163
reproducible features were detected respectively in the two systems with 26%
reproducibility obtained using both systems. Comparison of these features showed that a
proportion was only present in Jet Stream and the remainder were detectable at low
intensity and not reproducible.
324 Metabolomics

Fig. 2. Comparison of features observed with Jet Stream and ESI. Venn diagram showing
features found in human plasma extracts using LC 1290 coupled to 6530 QTOF coupled with
Jet Stream technology using sheath gas temperature of either 200°C or 400°C at 0.8 ml/min.
The features present at 200°C were 888, 1491 were present only at 400°C and 3438 were
present at both temperatures.
Improvement in the Number of Analytic Features Detected by Non-Targeted
Metabolomic Analysis: Influence of the Chromatographic System and the Ionization Technique 325

Fig. 3. Abundance of features observed exclusively at 200°C and 400°C. Heat plot depicting
the abundance of features present in human plasma extracts using LC1290 coupled to 6530
QTOF coupled with Jet Stream technology at either 200°C or 400°C at 0.8 ml/min. Each line
represents one feature found exclusively at either temperature, with red representing those
features present in a low high intensity and in blue those present in low intensity.

In summary, the Jet Stream technology increased the overall number of features when
compared with the ESI but thermal degradation occurred above 200°C, which is therefore
the optimal temperature to use under the conditions studied.
Our data demonstrates the advantage of the new LC system which allowed operation at
higher flow rates with low back pressure and very reproducible analysis. The increase in
flow rate resulted in a predictable increase in peak capacity (Figure 4).
326 Metabolomics

Fig. 4. Peak capacity versus flow rate determined for creatine (m/z 132.07), phenylalanine
(166.08), tryptophan (m/z 204.10) and glycerophosphocholine (m/z 496.34) and ion m/z
332.33 using the conventional 1200 LC system and novel 1290 LC system coupled to ESI and
Jet Stream technology. Chromatographic separation was carried out on a 100mmx2.1mm ID
reverse phase Waters Acquity T3 column with a 7.5min 0.1% formic acid: acetonitrile
gradient followed by 2 min isocratic.
However, saturation of desolvation may have occurred at flow rates of 0.8 ml/min and above,
as mentioned previously, and features started to disappear. For example we could no longer
detect creatine (m/z 132.07) and glycerophosphocholine (m/z 496.34) with ESI above a flow rate
of 0.6 ml/min. The Jet Stream technology detected these features at higher flow rate when
compared with the ESI and showed the expected linear increase in peak capacity with flow rate.
We were concerned however, that at higher flow rates, less data points were collected across
chromatographic peaks and proceeded to test various scan rates (2, 4 and 6Hz) at 0.6 and 0.8
ml/min (Table 4a). For a limited number of compounds the number of points across peaks
for early and late eluting metabolites (0.5-0.8 min, n=2, and 6-6.5 min, n=4) were measured).
Decreasing the scan rate increased the number of points across the peaks but decreased the
sensitivity of the analysis resulting in a significant decrease in the total and reproducible
number of features (Table 4b).
For example, at 0.8 ml/min, at 2Hz; only 4566 features were detected and 2056 with less than
25%CV as opposed to 3716 and 1294 at 4Hz and 3233 and 893 at 6Hz. At 2Hz, there were an
average number of 5 points across chromatographic peaks versus 15 at 4Hz, and in late eluting
peaks an average of 10 points were monitored across peaks at 2Hz and 25 at 4Hz.
Interestingly, the overall percentage of reproducible features was similar at all acquisition rates
suggesting that the loss of sensitivity observed at 4 or 6GHz occurs equally across total
features whether they are variable or not. The number of reproducible features before 2 min at
0.6 ml/min was: 28 at 2Hz versus 40 and 41 at 6Hz (data not shown). The lower number of
reproducible features at 2Hz when compared to 4 and 6 suggests that early polar metabolites
being defined with less than 10 points across peaks are less reproducibly detected.
Improvement in the Number of Analytic Features Detected by Non-Targeted
Metabolomic Analysis: Influence of the Chromatographic System and the Ionization Technique 327

2GHz 4GHz 6GHz 2GHz 4GHz 6GHz


m/z,RT 200°C 200°C 200°C m/z,RT 200°C 200°C 200°C
0.6mlmin 0.6mlmin 0.6mlmin 0.8mlmin 0.8mlmin 0.8mlmin
116.07_0.47 8 (7.5) 22 (5.4) 38 (6.7) 116.07_0.36 6 (4.5) 17 (5.5) 22 (5.2)
182.08_0.50 8 (3.8) 9 (1.3) 11 (2.7) 182.08_0.38 7 (4.0) 7 (1.6) 9 (3.1)
312.36_6.20 11 (0.4) 27 (0.4) 33 (0.2) 312.36_5.95 10 (0.5) 15 (1.0) 30 (0.8)
496.34_6.00 10 (2.4) 23 (2.5) 27 (2.5) 496.34_5.70 8 (1.6) 19 (2.0) 24 (2.6)
520.34_5.80 9 (1.5) 19 (1.0) 25 (1.3) 520.34_5.50 7 (1.6) 13 (1.60) 23 (0.9)
522.34_6.30 10 (1.0) 29 (1.0) 37 (0.8) 522.34_7.20 10 (0.8) 28 (0.8) 32 (0.5)
aChromatography performed on a reverse phase Waters Acquity T3 column with a 7.5min 0.1% formic

acid: acetonitrile gradient. Mass spectrometry analysis performed on 6530 QTOF in ESI mode.
Chromatograms were then analysed using MassHunter Qualitative software.
Table 4a. Evaluation of number of points across chromatographic peaks and mass accuracy
of selected metabolites using 2Hz, 4Hz and 6Hz scan rates at 0.6ml/min and 0.8ml/min a

Total no. of No. of features % of features


Method
features <25% CV with <25% CV
1290 Infinity LC 0.6ml/min 200°C 2HZ 4566 2056 45
1290 Infinity LC 0.6ml/min 200°C 4HZ 3058 1410 46
1290 Infinity LC 0.6ml/min 200°C 6HZ 2231 1073 48
1290 Infinity LC 0.8ml/min 200°C 2HZ 4460 2602 26
1290 Infinity LC 0.8ml/min 200°C 4HZ 3716 1294 35
1290 Infinity LC 0.8ml/min 200°C 6HZ 3233 893 28
aChromatography performed on a reverse phase Waters Acquity T3 column with a 7.5min 0.1% formic

acid: acetonitrile gradient. Mass spectrometry analysis performed on 6530 QTOF in ESI mode.
Chromatograms were then analysed using MassHunter Qualitative software.
Table 4b. The effect of scan rate on number and reproducibility of features present in
technical replicates of human plasma extracts the novel 1290 Infinity LC system coupled to a
6530 QTOF using Jet Stream technology a (n=3)

This study clearly demonstrates the considerable challenges associated with reproducibly
and sensitively acquiring metabolomic data. Increasing the flow rates eluted more non-polar
metabolites off the column but eventually at the detriment of polar metabolites that became
undersampled. Whatever choice is made in analytic conditions cannot be optimal for all
metabolites. It has to be noted that the number of features do not correspond to the number
of metabolites. For example other studies have described up to 23 features for a given
metabolite which further complicates matters (these may include multiply charged ions and
in source fragmentation ions) (Evans 2009).
In conclusion, our study describes the much improved effect of the 1290 LC system together
with the Jet Stream technology on the number of features detected compared with the 1200
LC system and ESI. It is clear that this increased number of features corresponds both to an
increased number of metabolites eluting from the column at higher flow rates and an
additional number of species being reproducibly ionized by the Jet Stream technology when
compared with the ESI source. Our preferred analytic conditions use the 100 mm analytic
column, a 7.5 min gradient (total run time 10 min plus 3 min equilibration) with a flow rate
of 0.6 ml/min and an acquisition rate of 2Hz.
328 Metabolomics

6. References
Brown, P. O.; Botstein, D. Nat Genet 1999, 21, 33-37.
Bruce S.J., Jonsson P., Clorarec C., Trygg J., Marklund S.L., Moritz T. Anal. Biochem., 2008,
372, 237-249.
Crews, B., Wikoff, W.R., Patti, G.J., Woo, H.K., Kalisiak, E., Heideker, J., and Siuzdak, G.
Anal Chem 2009, 81, 8538-8544.
Dunn, W.B. Phys Biol 2008, 5, 100-111
Evans, A.M., DeHaven C.D., Barrett T., Mitchell M. and Milgram E. J Anal Chem 2009
81 (16), pp 6656–6667
Kitano, H. Nature 2002, 420, 206-210.
Lai, L., Michopoulos, F., Gika, H., Theodoridis, G., Wilkinson, R.W., Odedra, R., Wingate, J.,
Bonner, R., Tate, S., and Wilson, I.D. Mol Biosyst 2010 6, 108-120.
Lee, S. H.; Woo, H. M.; Jung, B. H.; Lee, J.; Kwon, O. S.; Pyo, H. S.; Choi, M. H.; Chung, B. C.
Anal Chem 2007, 79, 6102-6110.
Lenz, E. M.; Bright, J.; Knight, R.; Wilson, I. D.; Major, H. Analyst 2004, 129, 535-541.
Nicholson, J. K.; Lindon, J. C. Nature 2008, 455, 1054-1056.
Nicholson, J. K.; Wilson, I. D. Nat Rev Drug Discov 2003, 2, 668-676.
Pandey, A.; Mann, M. Nature 2000, 405, 837-846.
Pandher, R., Ducruix, C., Eccles, S.A., and Raynaud, F.I. J Chromatogr B Analyt Technol
Biomed Life Sci. 2009, 877, 1352-1358.
Plumb, R.; Granger, J.; Stumpf, C.; Wilson, I. D.; Evans, J. A.; Lenz, E. M. Analyst 2003, 128,
819-823.
Quinones, M.P., and Kaddurah-Daouk, R. Neurobiol Dis 2009 35, 165- 176.
Sabatine M.S., Liu, E., Morrow, D.A., Heller E, McCarroll R., Wiegand R., Berriz G.F., Roth
F.P., Gerszten R.E., . Circulation. 2005, 112. 3868-3875.
Want E.J., O’Maille G., Smith C.A., Brabdon T.R., Uritboonthai W., Qui C., Trauger S.A.,
Siuzdak, G. Anal. Chem., 2006, 78, 743-752.
van der Greef, J.; Stroobant, P.; van der Heijden, R. Curr Opin Chem Biol 2004, 8, 559-565.
Zelena E., Dunn W, D. Broadhurst, S. Francis-McIntyreK. Carroll, P.Begley, S. O’Hagan, J.D.
Knowles, A. Halsall, HUSERMET Consortium, I.D. Wilson,D. Kell, D. Anal. Chem.,
2009, 81, 1357-1364.
Part 7

Metabolomics in Safety Assessments


14

Challenges for Metabolomics as a


Tool in Safety Assessments
George G. Harrigan and Bruce Chassy
Regulatory Product Characterization and Safety Center, Monsanto Company, St. Louis,
USA

1. Introduction
Agriculture’s ability to supply an abundance of nutritious foods and feeds to nourish the
world’s growing population faces serious challenges (Foresight, 2011). In order to meet
these challenges, plant breeders will be required to continuously improve agricultural
productivity as well as enhance food and feed quality. In recent years, the development of
methods for the direct introduction of new traits to produce transgenic varieties – also
known as GM crops – has proven to be a powerful tool in the hands of breeders. In most
countries, however, GM crops are subjected to rigorous pre-market regulatory assessments
that require numerous laboratory and field studies and which consume time and resources
(Kalaitzandonakes et al., 2007).
Comprehensive compositional analyses represent a key component of the pre-market safety
evaluations of GM crops (Harrigan, et al., 2010). These analyses typically include the
measurement of levels of key nutrients such as protein, storage oil, fiber, amino acids, fatty
acids, vitamins, as well as crop-specific metabolites such as gossypol and cyclopropenoid
fatty acids in cotton or isoflavones in soybean. The Organization of Economic Cooperation
and Development (OECD) has produced a series of consensus documents that identify key
analytes in a number of major crop varieties (http://www.oecd.org). These documents
carefully review the composition and uses for each crop and identify those components that
contribute to nutritional or functional food or feed value as well as components that might
confer health-beneficial, health-protective, or harmful effects (e.g. allergens, anti-nutrients,
and potential toxicants). The large-scale compositional studies performed as part of
regulatory assessments must follow internationally accepted guidelines. These are outlined
in detail by Codex Alimentarius (Codex Alimentarius, 2008) and OECD. In most cases, these
studies are typically conducted under Good Laboratories Practice (GLP), a practice that
places a high premium on documentation and reconstructability of data, method validation
and personnel training, and a requirement for professionally staffed Quality Assurance
Units.
The fact that different crops produce foods or feeds with differing compositions, along with
the fact that human and animal diets vary greatly in their consumption of these crops,
means that each crop plays a unique role in diet and health. Most plant foods in the human
diet make significant contributions to the total intake of just a few macro- and
332 Metabolomics

micronutrients (Senti and Rizek, 1974; Chassy, 2010). It is therefore important to assure that
no changes have occurred that would lower the dietary intake of an essential nutrient; on
the other hand, large changes in the content of one or more nutrients in a crop that supplies
an infrequently consumed food, one which is consumed in small amounts in the diet, or one
which is not an important source of that nutrient in the diet, are of no health consequence
and will have no adverse effect on health (Chassy, 2010).
The identification and analysis of a key set of relevant metabolites is often referred to a
“targeted” compositional analysis. Analyses utilize quantitative assays and the overall
approach allows the generation of data that is easily interpretable from a nutrition and
food/feed safety aspect. Furthermore, since the small molecule metabolite pool in seed is
of low abundance relative to macromolecular components, measurement of
macronutrients approximates the total seed biomass. For example, the small molecule
metabolite pool in corn grain is only ~5% of the total biomass (corn is dominated by
starch, fiber, protein, and fat). Anti-nutrient components in grain such as phytic acid and
raffinose (which represent much of the small molecule metabolite pool) are measured in
regulatory assessments. Other small molecules metabolites can be included if they are an
intended endpoint of compositional or nutritional modification. Otherwise analytical
measurement of the metabolites that constitute this pool, mainly ubiquitous free amino
acids, sugars, and organic acids), is of little value owing to the extreme sensitivity of
metabolite levels to environmental influences and the negligible contribution they make
to safety and nutritional content (Herman et al., 2009; Skogerson et al., 2010, Harrigan et
al., 2007).
In fact, levels of all crop compositional components are influenced markedly by
environment (Harrison and Harrigan, 2011; Harrigan, et al., 2010; Zhou et al., 2011a, 2011b).
To illustrate, as far back as 1983, it was noted that “The concentration of the isoflavones vary
from [soybean] variety to variety, and there are also differences when the same variety is
grown in different locations” (Eldridge and Kwolek, 1983). Given the extensive scientific
literature on isoflavone variability, it was unsurprising that Gutierrez-Gonzalez et al. (2009)
recently concluded that “The range of values of isoflavones is overwhelming, even for
homozygous genotypes growing in the same year and location, which greatly complicates
genetic studies.” This is true for almost all crop compositional components as evidenced by
challenges in enhancing nutritional quality in staple crops through conventional
approaches. Figure 1 illustrates the type of variability than can be observed for metabolites
such as isoflavones.
The use of multiple geographically separate sites is required in regulatory assessments to
allow compositional studies across a wide range of environmental conditions. Indeed,
information on compositional variation in conventional crops with respect to their
responsiveness to environmental factors is necessary to provide context to evaluations of
new GM crops. Studies incorporating four to five replicated field sites utilizing randomized
complete block designs with three blocks per comparator are typical in regulatory
assessments, although the European Food Safety Authority (EFSA) has recently mandated a
minimum of eight replicated sites utilizing randomized complete block designs with four
blocks (EFSA, 2011).
Challenges for Metabolomics as a Tool in Safety Assessments 333

Fig. 1. An overview of variability in isoflavone levels. Datapoints show daidzein values


from an analysis of GM (40-3-2) and conventional reference comparators from a total of nine
(2001-2009) growing seasons. A total of 112 unique GM varieties were assessed (Zhou et al.,
2011b). This type of information presents context to any GM-non-GM pairwise comparison
and would be a required component of any metabolomic assessments.

Results to date from these large-scale compositional studies have generally demonstrated
that the effect of transgene insertion is significantly less than the impact of environmental or
germplasm variation on conventional crops (Harrigan et al., 2010). This has allowed some to
question the relevance and design of compositional assessments. One review, for example,
suggests that “the current complexity and resource requirements for compositional studies
on transgenic crops containing input traits are not justified by a commensurate
understanding of safety” (Herman et al., 2009).
Despite continued confirmation that conventional breeding and environmental variation
contribute to compositional variability more so than transgene insertion (Ricroch et al.,
2011), and the resource-intensiveness of the large-scale studies currently required for
regulatory assessments, there remains some interest in the application of profiling
technologies to compare GM and conventional crops. These are often posited in terms of
“gap-filling” (Heineman et al., 2011) or “case-by-case” (Davies, 2010) evaluations. It is also
perceived by many (e.g., Kok et al., 2008) that measurement of “primarily the low-
molecular weight molecules” is more relevant to safety than proteomic or transcriptomic
profiling due to a closer relationship to “the plant phenotype and nutritional and
toxicological characteristics”. This potential advantage of metabolic profiling could be
extended as an improvement over, for example, measurements of gross levels of protein,
fat, and fibers, key nutritional but essentially “safe and inert” components of food. It is
334 Metabolomics

noteworthy that Kok et al. (2008) define metabolomics as the “generation of profiles of
secondary metabolites” whereas most metabolic profiling experiments to date have
focused on primary metabolites. It has also been suggested that untargeted profiling
techniques are unbiased while “targeted” compositional analysis is biased. Finally,
advocates of metabolomic profiling have suggested that such an approach can detect
potentially deleterious totally novel metabolites that would have been missed by
“targeted” analysis, although it should be noted that many profiling technologies require
standards of known identity to accurately identify and measure specific metabolites thus
limiting this potential advantage. In addition, in examples where a new traditionally bred
plant variety has caused toxic effects, this has been attributable to increased levels of well-
known toxicants (Chassy, 2010).
Profiling technologies have confirmed on a case-by-case basis the compositional
“equivalence” of GM crops to their conventional near-isogenic comparators (Ricroch et al.,
2011). Profiling technologies are, however, unlikely to provide immediately interpretable
data in safety assessments that would provide added value to, or otherwise enhance,
rigorously quantitative assessments of known nutrients and anti-nutrients that comprise
foodstuffs. In the case of metabolic profiling, this can be directly attributable to i) the
intrinsically safe nature of food itself, ii) inconsistencies in metabolite coverage versus
quantitative capabilities afforded by different data acquisition technologies, iii) the
ubiquitous and innocuous nature of small molecule metabolites identified in profiling as
well as extreme variability in metabolite levels even within homozygous genotypes, and iv)
the “chasm” between the large number of data generated in profiling experiments and the
ability to interpret them in a way that is meaningful to nutrition and food safety. We now
expand on these observations and further emphasize that a clear distinction between
“substantial equivalence” and food safety should be promoted.

2. Key challenges for the omics


1. Domesticated crops have been selected to serve human needs and have an extensive history of
safe consumption. Extensive information on levels of nutrients and crop-specific antinutrients is
available. These can be measured through highly quantitative assays to provide interpretable
data of direct relevance to food nutrition and safety.
Of over 250,000 plant species, only 7000 are considered as foodstuffs (Khoshbahkt and
Hammer, 2008), and even fewer, 150, supply over 90% of all plant food. Three major crops,
i.e. maize, wheat, and rice, supply over 66%. Choices made in crop domestication and
breeding have enabled food and feed qualities that serve human needs. Numerous path
changes between wild and domesticated plants are known and include e.g. loss of
spontaneous shattering of seed head on ripening and greater uniformity of seed ripening
and germination, both of which facilitate human harvest. A key path change however is
reduction and even loss of toxic compounds in the edible parts of domesticated crops. In
other words, human selection has resulted in crop phenotypes and compositions that
distinguish domesticated varieties from their natural counterparts, are more suited to
human diets and needs, and are safer and more nutritious. Interestingly, one review (Jones,
1998) asked “Why are so many food plants cyanogenic?” and concluded that “Cyanogenesis
by plants is not only a surprisingly effective chemical defence against casual herbivores, but
it is also easily overcome by careful pre-ingestion food processing, this latter skill being
Challenges for Metabolomics as a Tool in Safety Assessments 335

almost exclusive to humans.” In other words, because “cyanogenic plants are surprisingly
well protected from herbivory and yet can be readily detoxified by food processing, … early
farmers fortuitously chose these plants above all others for cultivation.”
Of course, many modern foodstuffs are still associated with “ancestral” secondary
metabolites that may confer nutritional or safety concerns at elevated levels. Classic
examples include glycoalkaloids in potato (NIEHS, 1998), β-N-oxalyl-L-α,β-
diaminopropionic acid (ODAP) in Lathyrus sativus (Bell, 2003), psoralens in celery (Beier and
Oertli, 1983), and gossypol in cotton (Sunlkumar et al., 2006). Targeted measurement of
these components as opposed to broad-based compositional screening is recommended by
Herman et al. (2009); in other words, compositional assessments should focus on molecules
explicitly associated with safety concerns. This is consistent with the observation that in the
very few examples where a new plant variety has caused toxic effects it has been
attributable to well-know toxicants associated with conventionally bred crops and not to a
hitherto undetected metabolite (Chassy, 2010).
It is noteworthy that such targeted assessments could easily facilitate a partnership with
omics researchers conducting semi-targeted profiling on pathways associated with toxic
metabolites to support both early development and commercialization of nutritionally
enhanced products. Such a partnership could, at least in principle, mitigate the current
regulatory burden imposed on new GM crops (Graff et al., 2009; Potrykus, 2010) and
promote the application of omics within modern agricultural biotechnology.
2. Information on compositional variation in conventional crops with respect to their
responsiveness to environmental and genetic factors is necessary to provide context to
evaluations of new GM crops. The need to assess natural variation is also true for metabolomics
yet little information on the impact of conventional breeding on metabolite profiles is available.
The inconsistent coverage of metabolites offered through different data acquisition platforms may
provide challenges in establishing a coherent literature in this area.
Ironically, as mentioned earlier, continued confirmation that conventional breeding,
environment, and germplasm contribute to compositional variation more than transgene
insertion has coincided with increased interest in the use of ‘omics technologies. This
paradox is compounded by the fact that results from these technologies have only
further highlighted the equivalence of GM crops to their conventional counterparts and
reaffirmed the substantial effect of environment and germplasm on compositional and
biochemical variability (see Ricroch et al., 2011). Although there are complexities in the
interpretation of data generated through modern profiling technologies (Broadhurst and
Kell, 2006; Lay et al., 2006) including the fact that the data is not quantitative and there is
no standardized framework for comparisons, the lack of variation between GM crops
and their conventional comparators at the transcriptomic, proteomic, and metabolomic
level has, nonetheless, been independently corroborated. These profiling evaluations
extend to a wide range of plants including wheat (Baker et al., 2006; Gregersen et al.,
2005; Ioset et al., 2007), potato (Catchpole et al., 2005; Defernez et al., 2004; Lehesranta et
al., 2005), soybean (Cheng et al., 2008), rice (Dubouzet et al., 2007; Wakasa et al., 2006),
tomato (Le Gall et al., 2003), tobacco, Arabidopsis (Kristensen et al., 2005), and Gerbera
(Ainasoja et al., 2008).
336 Metabolomics

As with the compositional studies reported above, results from many of the ‘omics studies
emphasize the need to understand natural variation in levels of endogenous metabolites in
providing biological context to pair-wise differences in any recorded profiles (see Figure 1).
Levels of compositional components are sensitive to environmental conditions. This has been
established for, for example, protein and oil in key crops (Panthee et al., 2005; Lam et al., 2010).
Protein levels in soybean seed generally average ~40% dry weight (dwt), with values reported
in the USDA soybean germplasm collection, for example, ranging from 34.1 to 56.8% dwt
(Wilson, 2004). In a recent meta-analysis of environmental effects on soybean composition,
Rotundo and Westgate (2009) observed that water stress, temperature, and/or nitrogen supply
all affected protein levels measured in mature seed.
Variability is even greater for lower abundance small molecule metabolites. Vitamin E (-
tocopherol) is typically only a minor component in soybean but is known to be important in
maintaining oxidative stability of soybean oil. Levels in soybean seed are affected by
environment and germplasm. For example, Britz et al. (2008) showed a greater than 2-fold
variation in levels across three locations in the U.S. over a period of four years. Levels in
soybean seed harvested from six different locations in Eastern Canada over a single year
ranged from 0.87 to 3.32 mg/100g dwt (Seguin et al. 2009). Seguin et al. (2010) point out that
environmental factors associated with variability in vitamin E levels include drought,
temperature, and even crop management systems. The “overwhelming variability” of
isoflavones was mentioned in the introduction (see Figure 1). As will be discussed later, this
“overwhelming variability” can be considered to apply to levels of small molecule
metabolites in harvested seed and grain of most crops.
Encouragingly, many comparative profiling studies on GM and non-GM crops have been
designed to include at least one element of genotypic or environmental variability. This is
exemplified in the following two examples, both of which reaffirm the need to provide
biological context to pairwise-differences between two comparators.
In Baker et al. (2006) NMR-based metabolic profiles of three GM wheat varieties and the
corresponding parents were generated. The incorporated transgenes encoded high-
molecular weight subunits of the storage protein, glutenin. The wheat varieties were grown
at two different sites over three different growing seasons (1999 -2001). Differences between
the GM and parental lines were within the same range as the differences between the
control lines grown on different sites and in different years. Analogous to the approach
adopted in targeted compositional analyses adopting OECD recommendations, this study
emphasized the importance of data from multiple years and multiple sites and that
environmental variation influences metabolome composition.
In Catchpole et al. (2005) two GM potato varieties modified in fructan chemistry were
grown over two different seasons (2001, 2003). Metabolic profiles of the GM and five
conventional crops were generated using flow-injection MS (FIE-MS), GC-MS, and LC-MS.
These demonstrated that differences between the GM and conventional potatoes were due
to the intended metabolic changes, but aside from these targeted changes, the GM crops
were “substantially equivalent to traditional cultivars”. A major finding recognized by the
authors was the large variation in the metabolic profiles of the conventional crops and, as
such, the study emphasized the importance of understanding genotypic variability in
assessments of compositional changes.
Challenges for Metabolomics as a Tool in Safety Assessments 337

An often overlooked aspect of the Catchpole et al. (2005) paper is their demonstration
that levels of glycoalkaloids (-chaconine and -solanine) were normal in the GM
potatoes, a result that is easily interpretable from a food and feed perspective. Our
understanding of nutrients and anti-nutrients forms the basis of attempt to modify crops
through conventional breeding or agricultural biotechnology. It has allowed crops to be
developed by conventional breeding that are deliberately non-equivalent to their
parental progenitors in a wide range of nutritional (and agronomic) characteristics.. As
Rischer et al. (2006) point out “For centuries, conventional plant breeding programs have
produced new traits, higher yields and improved quality. However, little attention has
been paid to metabolic changes occurring in successive generations. The issue has gained
importance only recently in the context of defining thresholds for safety assessments of
GM crops.” It is not immediately obvious why these hitherto neglected metabolites
should now be at the center of such attention. Indeed, there are few studies on small
molecule metabolite changes in crops where macro-molecular composition has been
deliberately changed through conventional breeding (e.g. high oil and high protein corn,
high oil soybean).
Catchpole et al. (2005) in their demonstration of the compositional equivalence of GM
potatoes to conventional lines also remark on the large metabolite variation in
conventional potato as follows; “These significant differences [between conventional
cultivars] were never sought as desired traits in traditional breeding programs, and
overall composition has not given cause for public safety concerns”. Overall, however,
experimental designs that will both account for natural variation and have enough power
to identify differences that can be attributed to transgene insertion will offer opportunities
to maximize the value of omics technologies as tools in plant breeding and the
development of new crops.
3. Metabolomics offers opportunities to generate data on large numbers of metabolites. Most of
these metabolites will be low in abundance and levels will be highly variable. They are also more
likely to include central (and hence ubiquitous) metabolites such as sugars, organic acids, and
free amino acids; metabolites that are not immediately associated with safety or nutritional
relevance.
Compositional assessments of new foodstuffs generally focus on the article of commerce,
most typically harvested seed or grain. This material is generally characterized by high
levels of starch, protein, fat, and fibers, with the small metabolite pool being low in
abundance. For example, approximately 95-98% of maize grain is comprised the
aforementioned materials; the small metabolite pool in grain, is of low abundance (~2-5% of
grain biomass) and its levels are highly dependent on changes in the macromolecular pool.
Soybean seed is comprised 40% protein, 20% fat, and 15% fiber. The residual 15% is
comprised mainly of sugars (e.g. sucrose, raffinose, stachyose, glucose, galactose, fructose)
of which the principal two, raffinose and stachyose, are measured in regulatory assessments.
The fact that the small molecule metabolite pool in seed or grain is of low abundance and
influenced by levels of the major macromolecular nutrients accounts for its extensive
variability (Skogerson et al., 2010; Harrigan et al., 2007).
Skogerson et al. (2010) sought to assess genetic and environmental impacts on the
metabolite composition of corn grain. Their data acquisition technology (gas
338 Metabolomics

chromatography-mass spectrometry) measured 119 identified metabolites including free


amino acids, free fatty acids, sugars, organic acids, and other small molecules in a range of
corn hybrids derived from 48 inbred lines crossed against two different tester lines (from the
C103 and Iodent heterotic groups) and grown at three locations in Iowa (Table 1). Different
metabolic phenotypes were clearly associated with the two distinct tester populations.
Overall, grain from the C103 lines contained higher levels of free fatty acids and organic
acids, whereas grain from the Iodent lines were associated with higher levels of amino acids
and carbohydrates. In addition, the fold-range of genotype mean values [composed of six
samples each (two tester crosses per inbred × three field sites)] for identified metabolites
ranged from 1.5- to 93-fold with sugars and polyols being particularly variable.
Interestingly, some grain metabolites showed a non-normal distribution over the entire corn
population, which could, at least in part, be attributed to large differences in metabolite
values within specific inbred crosses relative to other inbred sets.

Affected by
Metabolite class No. of analytes Affected by Testera
Locationb
free amino acids 26 14 2
sterols, amines, and
17 6 1
others
organic acids 17 6 0
free fatty acids and
17 5 0
related metabolites
sugar alcohols 18 5 0
mono-, di-, and
16 1 0
trisaccharides
sugar acids 8 0 1
aThis indicates a statistically significant difference (p<0.0001) between hybrids derived from a cross

with one tester (C103 heterotic group) versus another tester (Iodents heterotic group) bThis indicates a
statistically significant difference (p<0.0001) across the three locations in this study
Table 1. Variation in Metabolites due to Genotype or Environmental Variation

In an analogous report on the same samples, Harrigan et al. (2007) concluded that, given such
variability, measurement of the small metabolite pool, was unlikely to prove useful to a
comparative assessment of GM crops unless a given metabolite was an intended nutritional or
toxicological endpoint. In fact, it is not immediately obvious how the data generated in
Skogerson et al. (2011) could be used to determine which hybrids in this study were the safest.
In its report in 2004 the US National Research Council made pointed remarks about this
disconnect as summarized in the following quotes. “.. severe imbalances between highly
advanced analytical technologies and limited ability to interpret the results and predict
health effects that result from the consumption of food that is genetically modified” and
“….inherent difficulties, however, in identifying all of the constituents detected in profiling
methods or understanding the activity and potential biological consequence of all genes in
an organism severely limit the usefulness of these methods for predictive purposes..”
Unable to bridge this gap, many profiling proponents make an assumption of safety on the
non-GM comparator and consider statistical differences to equate with unintended effects.
This tendency is described later.
Challenges for Metabolomics as a Tool in Safety Assessments 339

4. Another challenge in establishing a coherent literature on the impact of conventional and other
approaches to breeding on natural variability in metabolite as well as determining a framework
to establish nutritional meaning from metabolite analysis is the differential coverage of
metabolites offered through the numerous data acquisition platforms available to omics
researchers.
As described in numerous articles on metabolomics, (e.g. Goodacre et al., 2004; Rischer and
Oksman-Caldentey, 2006; Kusano et al., 2011) the large physico-chemical diversity of small
molecule metabolites renders comprehensive metabolomic profiling through a single data
acquisition technology impossible. A range of technologies associated with different
detection capabilities (metabolite coverage and sensitivity), precision, resolution,
throughput and reproducibility are now extensively deployed by the research community.
Nuclear magnetic resonance spectroscopy (NMR), gas-chromatography mass spectrometry
(GC-MS), liquid-chromatography (LC)-MS utilizing different ionization modes, Fourier-
transform MS, and capillary electrophoresis (CE)- MS have all been applied in comparative
assessments of GM and non-GM crops. MS approaches predominate over NMR analyses
given their greater sensitivity and coverage; however this advantage does come at the
expense of quantitation (i.e. MS would need an internal standard for every metabolite to be
quantitated) and with a large number of unidentified MS signals in any metabolite profile.
Whilst it has been suggested that untargeted profiling techniques are unbiased, it is clear
that selection of a specific data acquisition technology is a bias and that this type of
analytical bias would need to be justified by pre-specified experimental hypotheses. This
justification would be critical in a Regulatory environment.
Recognizing inherent limitations for any given data acquisition technology Kusano et al. (2011)
applied a multi-platform approach to an evaluation of transgenic tomato. These authors used a
combination of GC-MS, LC-MS, and CE-MS with each technology covering distinct metabolite
classes. Free amino acids, sugars and organic acids were covered by GC-MS, larger molecules
(e.g. flavonoids) by LC-MS whereas CE-MS measured specific cations and anions. Overall, the
data generated 175 unique identified metabolites but a total of 1460 with “no or imprecise
metabolite annotation.” Of the identified metabolites, only 56 were observed in at least two
platforms. A total of 261 peaks showed no correlation with experimental factors (transgene,
cultivar, tissue type) and had to be removed from statistical analyses.
It is worth pointing out that two studies that assessed the metabolic profiles of grain from GM
maize containing the Cry1ab gene and that utilized the same data acquisition platform (NMR)
differed in their conclusions on the impact of transgene insertion on levels of free amino acids
(Manetti et al., 2006; Piccioni et al., 2009). Manetti et al. (2006) reported that the GM crop
included higher levels of sugars (glucose, sucrose, meliobiose), GABA, glutamine, and succinate
and decreased levels of alanine, asparagine, and choline. Piccioni et al. (2009) reported lower
levels of all amino acids, lower sugars, and lower succinate (and other organic acids). Piccioni et
al. (2009) were also able to report on metabolites observed in their NMR profiles but absent in
those of Manetti et al. (2006). Key design differences between the two studies include different
parental lines, different growth conditions, and sample extraction protocols.
Levandi et al. (2008) utilized CE-MS to compare levels of 27 metabolites in three different
GM maize lines also containing the Cry1ab gene. Some of these metabolites (e.g., certain free
amino acids, choline, GABA) were also recorded in the NMR platforms of Mannetti et al
340 Metabolomics

(2006) and Piccioni et al. (2009). No consistent association of these metabolites with the GM
trait when assessed over all three GM lines was observed, a conclusion in line with the
combined results of Mannetti et al. (2006) and Piccioni et al. (2009). Several of the
metabolites reported by Levandi et al. (2006) are more typically associated with other
taxonomic groupings, for example, graveolin (Ruta graveolus, Rutaceae) and lunarine,
(Lunaria annua, Brassicaceae). The assignment of peaks to metabolites not typically
associated with a genus or family would almost certainly require extensive validation in a
Regulatory environment.
Leon et al. (2009) utilized FT-MS on the same samples assessed by Levandi et al. (2008). This
allowed coverage of 5500 mass signals of which approximately 1000 could be assigned an
elemental composition. Those elemental compositions could be associated (through
MasstTRIX) with specific metabolic pathways (KEGG); these associations are referred to as
“isomeric hits”. This approach would identify any differences in GM and non-GM metabolic
profiles, especially where an elemental composition could be assigned, to be tentatively
associated with biochemical differences. Overall, it was shown that a greater number of
isomeric hits in pathways such as arachidonic acid metabolism, free amino acid metabolism,
purine metabolism, and folate biosynthesis were associated with the GM samples. A list of
33 possible compounds that could distinguish the GM and non-GM varieties was generated,
of which 12 could be confirmed in an orthogonal assay (CE-MS). The authors then indicated
that only four of these could be considered as potential GM “biomarkers”; L-carnitine,
apigenidin, 5, 6-dihydroxyindole, and one unidentified metabolite. There is little further
literature on levels of these metabolites in maize and the association of these metabolites as
GM biomarkers is almost certainly premature. Further, the interpretability of the Levandi et
al. (2009) approach is not at all clear; there are fewer isomeric hits associated with inositol
phosphate metabolism, yet levels of phytic acid have been well-established to be near-
identical in GM and non-GM maize. The association of isomeric hits for bile acid
biosynthesis, which is not typically associated with plant metabolism, is also difficult to
interpret.
In summary, different metabolic profiling platforms applied to similar biological questions
will yield non-overlapping solutions. This is due to differential metabolite coverage (even
within similar data acquisition technologies) and is compounded both by the number of
unidentified signals observed in current metabolite profiles and, in some cases,
“identification” of metabolites not previously known to be biosynthetically associated with
the plant species or genus in question.

3. Equating statistical equivalence with biosafety


Predetermined criteria would need to be established for any study protocol, data acquisition
steps or statistical analyses utilized in a safety assessment. As alluded to earlier, sampling
from multiple replicated field sites would be required. Discussions on the number of
replicates required to generate meaningful results from omics experiments are available as
well as on the potential for bias and over-fitting (Broadhurst and Kell, 2006; Goodacre et al.,
2007). Here we focus on the routine misinterpretation of “statistical significance” (Goodman,
2008) and the tendency to associate statistically significant differences between GM and a
non-GM comparator as, minimally, an unintended effect, and often to imply the statistical
difference raises a question about the safety of a newly evaluated crop.
Challenges for Metabolomics as a Tool in Safety Assessments 341

As indicated earlier, compositional assessments of GM crops involve direct comparisons of


levels of key nutrients and anti-nutrients in the new crop variety to those of a near-isogenic
conventional comparator. Statistical evaluations of the compositional data have typically
utilized classical frequentist significance testing. There are, however, several features of
significance hypothesis testing that impact its application to compositional comparisons
between crops with different agronomic qualities (Lecoutre, et al., 2001). Berger (1985), for
example, stated, “We know from the beginning that the point null hypothesis is almost
certainly not exactly true, and that this will always be confirmed by a large enough sample.
What we are really interested in determining is whether or not the null hypothesis is
approximately true.” There are many factors that impact crop composition, including
agronomic traits we seek to modify through plant breeding, (e.g. Scott et al., 2006; Uribelarrea et
al., 2004; Dornbos and Mullen, 1992; Hymowitz et al., 1972; Wilcox and Shibles, 2001; Yin
and Vyn, 2005) and any compositional changes that accompany enhanced agronomic
quality may confound interpretation of results generated through significance testing.
Statistical significance is used only as a first step in comparative assessments. The
interpretation of statistical significance from a p-value, the probability of an observed result or a
more extreme result occurring if the null hypothesis were true, does not imply biological
significance (Goodman, 2008). Statistically significant differences do not imply large
differences between GM and conventional comparators or that these comparators can be
easily distinguished from a biological perspective. In fact, the power of the experimental
designs (multiple highly replicated field trials) adopted in current compositional
assessments allows statistical significance to be assigned even where there are very small
difference in mean values of a given component but where the distribution of component
values overlap extensively. As such, significance approaches must be accompanied with
further data analysis encompassing discussion of magnitudes of differences, assessments of
component ranges, and the sensitivity of component values to environmental factors such as
location. This is consistent with the recommendation by Codex Alimentarius (2008, Ch. 44)
that “The statistical significance of any observed differences should be assessed in the
context of the range of natural variations for that parameter to determine its biological
significance.” It is further consistent with observations of high variability in crop
composition recorded in the scientific literature. The current scientific consensus is that, in
most if not all cases, statistically significant differences between GM and near-isogenic
conventional controls represent modest and nutritionally meaningless differences in
magnitude. For example, a recent review of studies on GM crop composition showed that
over 99% of all nutrient and antinutrients comparisons, where significant differences at the
5% level (=0.05) in mean values were observed, had a relative magnitude difference less
than 20%. These differences are considerably less than the range of values attributable to
germplasm and environmental factors (Harrigan et al., 2010).
Most metabolic profiling experiments utilize significance testing and Rischer and Oksman-
Caldentey (2006) refer to unintended effects as “effects which represent a statistically
significant difference (e.g. in chemical composition of the GM plant compared with a
suitable non-GM plant)” although they acknowledge that such differences would have to be
evaluated in the context of natural variability. One review that endorses the use of omics in
safety assessments suggests that “the amount of variation from genetic engineering should
be small (~3%).” (Heineman et al., 2011). Whilst this particular number is unrealistic since it
342 Metabolomics

falls well within the natural variability of metabolite levels and is even less than typical
experimental error, setting a universal threshold for relative magnitude of differences as a
trigger for further safety assessments of GM crops has been considered. In 2000, the Nordic
Council of Ministers recommended that if a component in a GM crop differed from the
conventional control by ±20% in relative magnitude, additional analyses of the GM crop
were warranted (cited in Hothorn and Oberdoerfer, 2006). This concept was refined to
account for the nutritional relevance of a component and the experimental precision of its
measurement (Hothorn and Oberdoerfer, 2006). Threshold ranges for GM components were
suggested as follows; 0.833-1.20 of the conventional control for “nutritionally very relevant”
components (minerals, vitamins, anti-nutrients, bioactives, essential amino acids, and fatty
acids), 0.769-1.30 for “relevant” (non-essential amino and fatty acids), and 0.667-1.50 for
components of “less relevance” (proximates, fiber). Suggestions for the use of limits and
triggers of this kind have been criticized for their failure to fully account for the role and
contributions of the specific crop in the human diet; and with GM crops in particular since
they are often not eaten as such but are used as a source of macronutrients such as oil, starch
and protein (Chassy, 2008; Chassy, 2010). As noted previously, most plant foods in the
human diet make significant contributions to the total intake of just a few macro- and
micronutrients and therefore even large compositional changes in a single crop plant might
produce little impact on the nutritional value of the overall diet. Chassy (2010) has observed
that composition cannot be viewed in isolation since the composition of the diet is far more
important than the composition of a single variety of a single crop. Strictly numerical
approaches have not been adopted in compositional studies and there is no reason they
would be relevant to profiling experiments.
At least one profiling study has attempted to apply statistical equivalence testing but again
falls prey to the dubious association of equivalence with safety. Kusano et al. (2011)
compared a GM-tomato (a miraculin protein expressor) to not only to the parental line but
to a panel of conventional reference varieties. The statistical design (described by the
authors as a proof-of-safety test) involved comparing the difference between test and control
and the determining whether these differences fell within equivalence limits established by
the reference varieties. However such a design makes more of a statement about the
selection of the reference substances and the control to which the GM-trait is introgressed,
and not about the effect of transgene insertion; the same test-to-control differences can be
equivalent or non-equivalent contingent on whether a limited or diverse range of genotypes
is available. The overall conclusion from the study however was that “miraculin over-
expressors are remarkably similar to the control line”.
In summary, there are no defined data analyses strategies currently being consistently
applied to profiling data that would facilitate interpretability of data.

4. Conclusion
There are clearly divergent views about the utility of ‘omics sciences in food safety
assessments. This paper has discussed some of the reasons metabolic profiling technologies
are, however, unlikely to provide immediately interpretable data in safety assessments that
would otherwise enhance rigorously quantitative assessments of known nutrients and anti-
nutrients that comprise foodstuffs. Indeed, it is not clear to the present authors that any new
types of data are in fact necessary to judge GM or other foods as safe. We are also unaware
Challenges for Metabolomics as a Tool in Safety Assessments 343

of any “gaps” in our compositional knowledge that might compromise safety and in fact,
our current understanding of plant anti-nutrients and toxicants, allows GM solutions to
enhancing food safety (e.g. Sunilkumar et al., 2006). The last 25 years of research on GM
plants and 15 years of commercial experience planting GM crops without harm or incident
suggest that no difference in safety that would require further analysis exists between GM
and crops bred by other strategies. All breeding induces genetic changes and these changes
give rise to transcriptomic, proteomic and metabolomic alterations.
We consider that metabolic profiling could increase its value in food safety science as well as
in the development of nutritionally enhanced crops as follows;
1. Improved compositional analysis. One potential target for future research could be to
develop metabolic screening methods that afford a comprehensive compositional
assessment in a single suite of determinations rapidly and at lower cost than traditional
targeted analysis. It is known that the metabolites in a cell form a large, complex and
interconnected network; one possible approach would be elucidation of key metabolic
compound whose determination might provide insight into the global concentrations of
numerous other metabolites. If such a validated analytical method could be developed
it would great aid research and development and would be particularly valuable in
assessments of nutritionally enhanced crops where changes in a specific pathway are
sought. However, metabolomic technologies are not able to supply this kind of analysis
and data.
2. Detection of novel toxicants. Targeted analysis is inherently incapable of assessing levels
of metabolites that are not selected (targeted) for analysis. Proponents of metabolic
profiling have argued that profiling might detect the emergence of previously unknown
novel toxicants presumably created by the breeding process. However, the abundance
of a few macro-components (protein, fiber, carbohydrate, lipids) and numerous minor
metabolites leaves little compositional “space” for novel toxicants. If wholly new
molecules were created by the spontaneous evolution of a new pathway or pathways
necessary for its biosynthesis, the chances that sufficient quantities would be present to
exert an adverse effect are small indeed. Perhaps this is why such effects have not yet
been observed by science or why coherent hypotheses as to how a novel toxicant would
be generated by a specific breeding process appear to be sparse in the literature.
3. Detection of unintended effects. Proponents of metabolic profiling often suggest that a
profile itself may be an indicator that unintended changes had occurred. Methods to
draw safety conclusions based on differences in metabolic profiles do not yet exist, and
certainly as we have discussed above, no reason to assume that differences in profiles
imply a safety concern; in fact, by any objective measure, there is no such technique as
metabolomic profiling. What we have today is a series of distinct and emerging
powerful scanning techniques each of which surveys a slightly different molecular
landscape with variable degrees of resolution. Clearly, the number of metabolites
present in crops is very large and the power of targeted metabolic profiling will become
increasingly useful in analyzing the chemical complexity of prospective commercial
releases as they progress through initial research and development phases.
Metabolomics is an expanding and exciting field of research. The rapidly expanding scope
of the metabolomic profiling technologies tempts us to test their applicability to a wide
344 Metabolomics

array of analytical challenges. We have, on the other hand, a long history of safe experience
with plant breeding. We know that many unintended changes take place in plant breeding,
however, these are almost without exception innocuous. There is no reason to believe that
GM breeding should require any new or different data set than other forms of breeding.
It seems clear to the present authors that there is no role for metabolic profiling in food
safety assessment. We agree that modern targeted metabolic profiling technologies can
rapidly identify pathway perturbations and, if judiciously applied and interpreted, might
enhance food safety science, although traditional analytical methods can still be used to
assess if changes in pathways and metabolite pools have occurred. If incorporated into the
early selection stages of a prospective new trait targeted metabolic profiling may greatly aid
in the selection of metabolites that need to be considered during the compositional phase of
a risk assessment. To quote Larkin and Harrigan (2007) “However, it should be self-evident
that GM crops ought not to be considered a single monolithic class that is either good or bad
for the economy, agriculture or the environment. Each novel crop should be considered on
its own merits and demerits. If we ever get to that point we will have achieved something
positive out of the GM controversy.” It is our hope that colleagues will take this as a
challenge to further metabolic profiling in the advancement of food safety and nutritional
enhancement of crops.

5. Acknowledgements
Figure 1 was prepared by Jay Harrison of the Statistics Technology Center, Monsanto
Company.

6. References
Ainasoja, M. M., Pohjala, L. L., Tammela, P.S. M., Somervuo, P. J., Vuorela, P. M. & Teeri, T.
H. 2008. Comparison of transgenic Gerbera hybrida lines and traditional varieties
shows no differences in cytotoxicity or metabolic fingerprints. Transgenic Res, 17,
793-803.
Baker, J. M. Hawkins, N. D. Ward, J. L., Lovegrove, A., Napier, J. A., Shewry, P. R., & Beale,
M. H. 2006. A metabolomic study of substantial equivalence of field-grown
genetically modified wheat. Plant Biotechnol J, 4, 381-92.
Beier, R. C. & Oertli, E. H. 1983. Psoralen and other linear furocoumarins as phytoalexins in
celery. Phytochem, 22, 2595-97.
Bell, A. E. 2003. Nonprotein amino acids of plants: Significance in medicine, nutrition, and
agriculture. J Agric Food Chem, 51, 2854–65.
Berger, J.O. 1985. Statistical Decision Theory and Bayesian Analysis. 2nd edition: Springer-
Verlag.
Britz, S. J., Kremer, D. F., & Kenworthy, W. 2008. Tocopherols in soybean seeds; Genetic
variation and environmental effects in field-grown crops. J Am Oil Chem Soc, 85,
931-36.
Broadhurst, D. I. & Kell, D. B. 2006. Statistical strategies for avoiding false discoveries in
metabolomics and related experiments. Metabolomics, 2, 171-96.
Catchpole, G. S., Beckmann, M., Enot, D. P., Mondhe, M., Zywicki, B., Taylor, J., Hardy, N.,
Smith, A., King, R. D., Kell, D. B., Fiehn, O. & Draper, J. 2005. Hierarchical
Challenges for Metabolomics as a Tool in Safety Assessments 345

metabolomics demonstrates substantial compositional similarity between


genetically modified and conventional potato crops. Proc Natl Acad Sci U S A, 102,
14458-62.
Chassy, B., Egnin, M., Gao, Y., Glenn, K., Kleter, G. A., Nestel, P., Newell-Mcgloughlin, M.,
Phipps, R. H. & Shillito, R. 2008. Recent developments in the safety and nutritional
assessment of nutritionally improved foods and feeds. Comp Rev Food Sci Food
Safety, 7, 50-113.
Chassy, B. 2010. Can ‘omics inform a food safety assessment? Reg Toxicol Pharmacol,58,
S62-70.
Cheng, K. C., Beaulieu, J., Iquira, E., Belzile, F. J., Fortin, M. G., & Stromvik, M. V. 2008.
Effect of transgenes on global gene expression in soybean is within the natural
range of variation of conventional cultivars. J Agric Food Chem, 56, 3057–67.
Codex Alimentarius. 2008. Guideline for the conduct of food safety assessment of foods
derived from recombinant-DNA plants. CAC/GL 45-2003.
Davies, H. 2010. A role for “omics” technologies in food safety assessment. Food Control, 21,
1601-10.
Defernez, M., Gunning, Y. M., Parr, A. J., Shepherd, L. V., Davies, H. V. & Colquhoun, I. J.
2004. NMR and HPLC-UV profiling of potatoes with genetic modifications to
metabolic pathways. J Agric Food Chem, 52, 6075-85.
Dornbos, D. & Mullen, R. 1992. Soybean seed protein and oil contents and fatty acid
composition adjustments by drought and temperature. J Am Oil Chem Soc, 69, 228-
31.
Dubouzet, J. G., Ishihara, A., Matsuda, F., Miyagawa, H., Iwata, H. & Wakasa, K. 2007.
Integrated metabolomic and transcriptomic analyses of high-tryptophan rice
expressing a mutant anthranilate synthase alpha subunit. J Exp Bot 58, 3309-21.
Eldridge, A. C. & Kwolek W. F. 1983. Soybean isoflavones: effect of environment and variety
on composition. J Agric Food Chem, 31, 394-6.
EFSA. 2011. EFSA Panel on Genetically Modified Organisms (GMO); Scientific opinion on
guidance for risk assessment of food and feed from genetically modified plants.
EFSA Journal, 9, 2150 [37pp]. Available online:
www.efsa.europa.eu/efsajournal.htm
Foresight. The Future of Food and Farming (2011) Final Project Report. The Government
Office for Science, London.
Goodacre, R., Vaidyanathan, S., Dunn, W. R., Harrigan, G. G. & Kell, D. B. 2004.
Metabolomics by numbers-Acquiring and understanding global metabolite data.
Trends Biotechnol, 22, 245-52.
Goodacre, R., Broadhurst, D., Smilde, A. K., Kristal, B.S., Baker, D. J., Beger, R., Bessant, C.,
Connor, S., Capuani , G., Craig, A., Ebbels, T., Kell, D. B., Manetti, C., Newton, J.,
Paternostro, G., Somorjai, R., Sjostrom, M., Trygg, J. & Wulfert, F. 2007. Proposed
minimum reporting standards for data analysis in metabolomics. Metabolomics, 3,
231-41.
Goodman, S. 2008. A dirty dozen: Twelve p-value misconceptions. Sem Hematol, 45, 135-40.
Graff, G. D., Zilberman, D. & Bennett, A. B. 2009. The contraction of agbiotech product
quality innovation. Nat Biotechnol, 27, 702-4.
346 Metabolomics

Gregersen, P. L., Brinch-Pedersen, H. & Holm, P. B. 2005. A microarray-based comparative


analysis of gene expression profiles during grain development in transgenic and
wild type wheat. Transgenic Res, 14, 887-905.
Gutierrez-Gonzalez, J. J., Wu, X., Zhang, J., Lee, J. D., Ellersieck, M., Shannon, J. G., Yu, O.,
Nguyen, H. T. & Sleper, D.A. 2009. Genetic control of soybean seed isoflavone
content: importance of statistical model and epistasis in complex traits. Theor Appl
Genet, 119, 1069-83.
Harrigan, G. G. & Harrison, J. M. 2011. Assessing compositional variability through
graphical analysis and Bayesian statistical approaches; case studies on transgenic
crops. Biotechnol Genet Eng Rev, 28, 1-18.
Harrigan, G. G., Stork, L. G., Riordan, S. G., Reynolds, T. L., Ridley, W. P., Masucci, J. D.,
Macisaac, S., Halls, S. C., Orth, R., Smith, R. G., Wen, L., Brown, W. E., Welsch, M.,
Riley, R., Mcfarland, D., Pandravada, A. & Glenn, K. C. 2007. Impact of genetics
and environment on nutritional and metabolite components of maize grain. J Agric
Food Chem, 55, 6177-85.
Harrigan, G. G., Lundry, D., Drury, S., Berman, K., Riordan, S. G., Nemeth, M. A., Ridley,
W. P. & Glenn, K. C. 2010. Natural variation in crop composition and the impact of
transgenesis. Nat Biotechnol, 28, 402-4.
Heineman, J. A., Kurenbach, B. & Quist, D. 2011. Molecular profiling - a tool for addressing
emerging gaps in the comparative risk assessment of GMOs. Environ Int, 37, 1285-
93.
Herman, R. A., Chassy, B. M. & Parrott, W. 2009. Compositional assessment of transgenic
crops: an idea whose time has passed. Trends Biotechnol, 27, 555-7.
Hothorn, L. A. & Oberdoerfer, R. 2006. Statistical analysis used in the nutritional assessment
of novel food using the proof of safety. Reg Toxicol Pharmacol, 44, 125-135.
Hymowitz, T., Collins, F. I., Panczner, J. & Walker, W. M. 1972. Relationship between the
content of oil, protein, and sugar in soybean seed. Agron J, 64, 613-16.
Ioset, J. R., Urbaniak, B., Ndjoko-Ioset, K., Wirth, J., Martin, F., Gruissem, W., Hostettmann,
K. & Sautter, C. 2007. Flavonoid profiling among wild type and related GM wheat
varieties. Plant Mol Biol, 65, 645-54.
Kok, E. J., Keijer, J., Kleter, G. A. & Kuiper, H. A. 2008. Comparative safety assessment of
plant-derived foods. Regul Toxicol Pharmacol, 50, 98-113.
Kristensen, C. Morant M., Olsen C. E., Ekstrøm, C. T., Galbraith, D. W., Møller, B. L., & Bak,
S. 2005. Metabolic engineering of dhurrin in transgenic Arabidopsis plants with
marginal inadvertent effects on the metabolome and transcriptome. Proc Natl Acad
Sci U S A, 102, 1779-84.
Jones, D.A. 1998. Why are so many food plants cyanogenic? Phytochem, 47, 155-62.
Kalaitzandonakes, N., Alston, J.M. & Bradford, K. J. 2007. Compliance costs for regulatory
approval of new biotech crops. Nat Biotechnol, 25, 509-11.
Khoshbahkt, K. & Hammer, K. 2008. How many plant species are cultivated? Genet Resour
Crop Evol, 55, 925-28
Kusano, M., Redestig, H., Hirai, T., Oikawa, A., Matsuda, F., Fukushima, A., Arita, M.,
Watanabe, S., Yano, M., Hiwasa-Tanase, K., Ezura, H. & Saito, K. 2011. Covering
chemical diversity of genetically-modified tomatoes using metabolomics for
objective substantial equivalence assessment. PLoS, 6, e16989.
Challenges for Metabolomics as a Tool in Safety Assessments 347

Lam, H-M., Xu, X., Liu, X., Chen, W., Yang, G., Wong F. L., Li, M.-W., He, W., Qin, N.,
Wang, B., Li, J., Jian, M., Wang, J., Shao, G., Wang, J., Sun , S. S.-M., & Zhang, G.
2010. Resequencing of 31 wild and cultivated soybean genomes identifies patterns
of genetic diversity and selection. Nature Gen, 42, 1053-59.
Larkin, P. & Harrigan, G. G. 2007. Opportunities and surprises in crops modified by
transgenic technology: metabolic engineering of benzylisoquinoline alkaloid,
gossypol and lysine biosynthetic pathways. Metabolomics, 3, 371-82.
Lay Jr., J.O, Borgmann, S., Liyanage, R. & Wilkins, C.L. 2006. Problems with the ‘‘omics’’.
Trends Anal Chem, 25, 1046-56.
Le Gall, G., Colquhoun, I. J., Davis, A. L., Collins, G. J. & Verhoeyen, M. E. 2003. Metabolite
profiling of tomato (Lycopersicon esculentum) using 1H NMR spectroscopy as a tool
to detect potential unintended effects following a genetic modification. J Agric
Food Chem, 51, 2447-56.
Lecoutre, B., Lecoutre, M.-P. & J. Poitevineau, J. 2001. Uses, abuses and misuses of
significance tests in the scientific community: Won't the Bayesian choice be
unavoidable? Int Stat Rev, 69, 399-417.
Lehesranta, S. J., Davies, H. V., Shepherd, L. V., Nunan, N., Mcnicol, J. W., Auriola, S.,
Koistinen, K. M., Suomalainen, S., Kokko, H. I. & Karenlampi, S. O. 2005.
Comparison of tuber proteomes of potato varieties, landraces, and genetically
modified lines. Plant Physiol, 138, 1690-9.
Leon, C., Rodriguez-Meizoso, I., Lucio, M., Garcia-Canas, V., Ibanez, E., Schmitt-Kopplin, P.
& Cifuentes, A. 2009. Metabolomics of transgenic maize combining Fourier
transform-ion cyclotron resonance-mass spectrometry, capillary electrophoresis-
mass spectrometry and pressurized liquid extraction. J Chromatog A, 1216, 7314-
23.
Levandi, T., Leon, C., Kaljurand, M., Garcia-Canas, V. & Cifuentes, A. 2008. Capillary
electrophoresis time-of-flight mass spectrometry for comparative metabolomics of
transgenic versus conventional maize. Anal Chem, 80, 6329-35.
Manetti, C., Bianchetti, C., Casciani, L., Castro, C., Di Cocco, M. E., Miccheli, A., Motto, M. &
Conti, F. 2006. A metabonomic study of transgenic maize (Zea mays) seeds revealed
variations in osmolytes and branched amino acids. J Exp Bot, 57, 2613-25.
NIEHS. 1998. National Institute of Environmental Health Sciences; -Chaconine [20562-03-2]
and -solanine [20562-02-1]. Review of toxicological literature. http://ntp-
server.niehs.nih.gov/index.cfm?objectid=6F5E930D-F1F6-975E-
7037ACA48ABB25F4
NRC. 2004. National Research Council: Safety of Genetically Engineered Foods. Approaches
to Assessing Unintended Health Effects. The National Academies Press,
Washington, D.C.
Panthee, D. R., Pantalone, V. R., West, D. R., Saxton A. M., & Sams, C. E. 2005. Quantitative
trait loci for seed protein and oil concentration and seed size in soybean. Crop Sci,
45, 2015-22.
Piccioni, F., Capitani, D., Zolla, L. & Mannina, L. 2009. NMR metabolic profiling of
transgenic maize with the Cry1A(b) gene. J Agric Food Chem, 57, 6041-49.
Potrykus, I. 2010. Regulation must be revolutionized. Nature, 466, 561
348 Metabolomics

Ricroch, A. E., Berge, J. B. & Kuntz, M. 2011. Evaluation of genetically engineered crops
using transcriptomic, proteomic, and metabolomic profiling techniques. Plant
Physiol, 155, 1752-61.
Rischer, H. & Oksman-Caldentey, K.-M. 2006. Unintended effects in genetically modified
crops: revealed by metabolomics? Trends Biotechnol, 24, 102-4.
Rotundo, J.L. & Westgate, M. E. 2009. Meta-analysis of environmental effects on soybean
seed composition. Field Crops Res, 110, 147-56.
Scott, M. P., Edwards, J. W., Bell, C. P., Schussler, J. R. & Smith, J. S. 2006. Grain composition
and amino acid content in maize cultivars representing 80 Years of commercial
maize varieties. Maydica, 51, 417-23.
Seguin, P., Turcotte, P., Tremblay, G., Pageau, D., & Liu, W. 2009. Tocopherols concentration
and stability in early maturing soybean genotypes. Agron. J, 101, 1153-59.
Seguin, P., Tremblay, G., Pageau, D. & Liu, W. 2010. Soybean tocopherol concentrations are
affected by crop management. J Agric Food Chem, 58, 5495-5501.
Senti, F.R. & Rizek, R. R.1974. An overview of GRAS regulations and the effect from the
viewpoint of nutrition, In Hanson C. H. (ed) CSSA Special Publication. Vol. 5. SSA,
Madison, WI.
Skogerson, K., Harrigan, G. G., Reynolds, T. L., Halls, S. C., Ruebelt, M., Iandolino, A.,
Pandravada, A., Glenn, K. C. & Fiehn, O. 2010. Impact of genetics and environment
on the metabolite composition of maize grain. J Agric Food Chem, 58, 3600-10.
Sunilkumar, G., Campbell, L. M., Puckhaber, L., Stipanovic, R. D. & Rathore, K. S. 2006.
Engineering cottonseed for use in human nutrition by tissue-specific reduction of
toxic gossypol. Proc Natl Acad Sci U S A, 103, 18054-59.
Uribelarrea, M., Below, F. E. & Moose, S.P. 2004. Grain composition and productivity of
maize hybrids derived from the Illinois protein strains in response to variable
nitrogen supply. Crop Sci, 44, 1593-1600.
Yin, X.H. & Vyn, T. J. 2005. Relationships of isoflavone, oil, and protein in seed with yield of
soybean. Agron J, 97, 1314-21.
Wakasa, K., Hasegawa, H., Nemoto, H., Matsuda, F., Miyazawa, H., Tozawa, Y., Morino, K.,
Komatsu, A., Yamada, T., Terakawa, T. & Miyagawa, H. 2006. High-level
tryptophan accumulation in seeds of transgenic rice and its limited effects on
agronomic traits and seed metabolite profile. J Exp Bot, 57, 3069-78 .
Wilcox, J.R. & Shibles, R. M. 2001. Interrelationships among seed quality attributes in
soybean. Crop Sci, 41, 11-4.
Wilson, R. F. 2004. Seed composition, In H.R. Boerma and J.E. Specht (ed.) Soybeans:
Improvement, production and uses, 3rd ed. ASA, CSSA, SSA, Madison, WI.
Zhou, J., Berman, K. H., Breeze, M. L., Nemeth, M. A., Oliveira, W. S., Braga, D. V., Berger,
G. U. & Harrigan, G. G. 2011a. Compositional variability in conventional and
glyphosate-tolerant soybean varieties grown in different regions in Brazil. J Agric
Food Chem, 59, online
Zhou, J., Harrigan, G. G., Berman, K. H., Webb, E. G., Klusmeyer, T. H. & Nemeth, M. A.
2011b. Stability of the compositional equivalence of grain from insect-protected
corn and seed from herbicide-tolerant soybean over multiple seasons, locations and
breeding germplasms. J Agric Food Chem, 59, 8822-28.
15

Metabolomics Approach for Hazard


Identification in Human Health Assessment of
Environmental Chemicals
Suryanarayana V. Vulimiri1, Brian Pachkowski2,
Ambuja S. Bale1 and Babasaheb Sonawane1
1U.S. Environmental Protection Agency, Office of Research and Development,
2Oak Ridge Institute for Science and Education Postdoctoral Fellow,

National Center for Environmental Assessment, Washington DC,


USA

1. Introduction
Exposure to xenobiotics induces complex biochemical responses in mammalian cells
resulting in several perturbations in cellular toxicity pathways. Within the context of
systems biology, such biochemical perturbations can be studied individually using “omics”
approaches such as toxicogenomics, transcriptomics, proteomics and metabolomics (Heijne
et al., 2005). The objective of this chapter is to examine how the metabolomics approach can
be used in identifying the risk posed by environmental chemicals to human health using
selective examples of organ toxicity. Metabolomics is a medium-to-high throughput
technique employing predominantly mass spectrometry (MS) and nuclear magnetic
resonance (NMR) technology (Roux et al., 2011) for the identification and characterization of
endogenous metabolites of low molecular weight (<1800 Da) arising from different
biochemical pathways either as primary or secondary metabolites (Idle & Gonzalez, 2007).
The sum total of all small metabolites is referred to as the “metabolome”. Metabolomics has
also been applied to the identification of low molecular weight, exogenous metabolites of
xenobiotics (Roux et al., 2011; Rubino et al., 2009). With these capabilities, metabolomics
represents a relatively quick and informative approach for assessing the physiological
response to environmental chemicals.

2. Human health risk assessment


Chemicals in the environment could pose potential risks to human health. In order to inform
the assessment of risks from chemical exposures, the U.S. National Research Council (NRC)
published a report entitled, “Risk Assessment in the Federal Government: Managing the Process,”
more commonly known as the “Red Book” (NRC, 1983), which has been widely accepted
and endorsed by the U.S. Environmental Protection Agency (U.S. EPA) and other federal
agencies. This risk assessment process consists of four steps: hazard identification, dose-
response assessment, exposure assessment, and risk characterization. The focus herein is on
hazard identification, which has been defined as “identification of the contaminants that are
350 Metabolomics

suspected to pose health hazards, quantification of the concentrations at which they are
present in the environment, a description of the specific forms of toxicity (i.e. neurotoxicity,
carcinogenicity, etc.) that can be caused by the contaminants of concern, and an evaluation
of the conditions under which these forms of toxicity might be expressed in exposed
humans” (NRC, 1994).
For human health assessment of chemicals, non-cancer or cancer risk values are derived
based on the selection of a critical endpoint of toxicity or several endpoints (e.g. biochemical,
pathological, physiological, and behavioral abnormalities) of adverse health outcomes.
Uncertainty factors are applied to the lowest dose associated with the critical health
outcome(s) in order to derive the resulting exposure level for non-cancer toxicity. These
uncertainty factors attempt to account for exposure duration, pharmacokinetic, and
pharmacodynamic data gaps associated with inter- and intra-species extrapolation.
The U.S. EPA and the International Agency for Research on Cancer (IARC) evaluate the
evidence for carcinogenesis in humans from epidemiological, experimental animal, and
mechanistic data to determine the qualitative cancer classification for humans. In addition,
the U.S. EPA evaluates exposure-response relationships and develops quantitative cancer
risk values based on the observed tumors that correspond to a unit exposure (U.S. EPA,
2005). Uncertainties with cancer risk values are presented and are generally associated with
the mode of action (MOA) for carcinogenicity.
One of the major concerns with cancer risk assessment is false-positive animal tumor
findings. Having an understanding of the mechanism(s) leading to carcinogenicity would
help in developing a better perspective of whether a carcinogen in experimental animals is
likely to be a carcinogen in humans. For example, correlating a metabolomic profile of a
suspected carcinogen between human exposures (environmental or occupational) and
experimental animal exposure studies would be highly useful. If similar biochemical
markers were to appear across the human and animal metabolomic profiles, that
information would help in informing similarities or differences in interspecies mechanisms.
Further, if the chemical was demonstrated to be a carcinogen in animals through a
traditional two-year animal bioassay, but there was inconclusive epidemiological evidence,
the similarity in metabolomic data could be used along with other mechanistic data (e.g.
mutagenicity/genotoxicity assays, cell proliferation findings, oxidative stress, epigenetics,
etc.) to support or refute human carcinogenicity. In this regard, metabolomics information
could be used to support mechanistic data to augment the animal and human findings.

3. The potential of “omics” data to inform mode of action of environmental


chemicals
In developing a human health evaluation for environmental chemical hazard
identification, it is ideal to have information on the key mechanistic events leading to an
adverse health outcome. In this regard, mode of action (MOA) is an important part of
hazard identification. MOA can be defined as “a sequence of key events and processes,
starting with interaction of an agent with a cell, proceeding through operational and
anatomical changes, and resulting in cancer formation” (U.S. EPA, 2005). A ‘key event’ is
defined as “an empirically observable precursor which is by itself a necessary component
of the MOA or is a biologically-based measurable marker for such a component” (U.S.
Metabolomics Approach for Hazard Identification in
Human Health Assessment of Environmental Chemicals 351

EPA, 2005). In vitro systems (e.g. cell cultures) and in vivo models (e.g. experimental
animals or human population studies) have identified several of the early key events
such as oxidative stress, inflammation, genotoxicity, and cytotoxicity that occur from
toxicant exposure. Since metabolomics measures biological response at the molecular
level, this approach can identify the metabolites associated with the sequence of ‘key
events’ and the processes inherent to the mechanism(s) of xenobiotic toxicity. The
metabolomics approach could generate several mode(s) of action hypotheses using a
nontargeted approach. The individual MOA hypothesis thus generated could be tested
in targeted approaches (e.g. measuring glutathione reduction from oxidative stress)
using more conventional assays. Metabolomics data could be used to further inform the
mode(s) of action in experimental animals associated with carcinogenicity or with non-
cancer health outcomes, which may help to confirm the relevancy of the observations in
experimental animals to humans.

4. Metabolomics approach in investigating environmental chemical exposure


Environmental chemicals act through multiple toxicity pathways via a multitude of
mechanisms (Guyton et al., 2009). To date, very limited toxicogenomics information has
been applied to the field of risk assessment (Boverhof & Zacharewski, 2006; Mortensen &
Euling, 2011). There is a paucity of relevant metabolomic information for application to
human health risk assessment of environmental chemicals. To date, the available literature
suggests an informative role of metabolomics in understanding the mode of action of
environmental xenobiotics (Vulimiri et al., 2011; Vulimiri et al., 2009).
In general, human data are relatively sparse for many environmental chemicals with respect
to both non-cancer and cancer health outcomes. Most human data are occupational, where
exposure levels are generally higher than those encountered in the environment. In many
cases, there are not any environmental or occupational human exposures that could be used
to corroborate the animal data. In these cases, animal data are generally used to develop
non-cancer and cancer risk values for human health assessments thereby raising issues of
uncertainty associated with interspecies extrapolation. Metabolomic data could be used to
fill in such data gaps. For example, in vitro metabolomic assays with human cells may be
developed to compare with animal metabolomic profiles to determine if there are
potentially similar mechanisms of toxicity or for identifying toxicity pathways. As a result,
this characterization of biochemical mechanisms of toxicity would inform hazard
identification for use in the human health risk assessment process.

5. Ability of metabolomics to differentiate gender, phenotypic, and genetic


differences, and organ-specific effects
Since metabolomics analyzes endogenous and exogenous (xenobiotic-derived) low
molecular weight metabolites, this approach has been applied to the differentiation of
metabolic profiles between phenotypes and genotypes. As briefly discussed below,
metabolomics has the ability to inform gender, genetic, and phenotypic differences as well
as organ-specific effects. For understanding the toxicity of environmental agents, the utility
of such information would clarify toxicodynamic uncertainty associated with the
extrapolation between species as well as within species (i.e. human) variability.
352 Metabolomics

5.1 Gender differences


Metabolomics approach can differentiate between gender-dependent differences in the
metabolic profiles of untreated or control experimental animals. In a study aimed at the
identification of novel biomarkers of effect using chemicals with diverse modes of action,
one research group (van Ravenzwaay et al., 2007) pooled the metabolomic data (i.e. plasma
profiles) from eleven individual experiments involving 670 male and female control Wistar
rats over a span of one year. After principal component analysis (PCA), the authors found
that the metabolic profiles were clustered into separate groups for males and females
suggesting a stable metabolome of the control rats during the study period. This observation
highlights the ability of metabolomics to potentially identify unique gender responses to
chemical exposure.

5.2 Genetic differences and phenotypic effects


The metabolomics approach can be further used for studying the relationship between the
genotype and phenotype of the organism. Genetic polymorphisms in human genes are
known to modify exposure to environmental health hazards and are a source of
uncertainty when assessing risk from environmental chemicals (Ginsberg et al., 2009;
Kelada et al., 2003). Genetic differences have been shown to reflect changes in the
metabolite profiles of individuals. In a human population, genetically determined variants
(e.g. those associated with fatty acid metabolism) in metabolic phenotype (metabotype)
have been identified by simultaneously detecting single-nucleotide polymorphisms
(SNPs) in a genome-wide association study (GWAS) and endogenous serum metabolites
(Gieger et al., 2008).
This is also evident in the field of functional genomics where a change in phenotype is
observed due to gene-related alterations (reverse-genetics) such as deletions or insertions
leading to silent mutations as in yeasts (Raamsdonk et al., 2001). Specific gene mutations can
also be evaluated using metabolic footprinting (Szeto et al., 2010). Such metabolomic data
could provide basic information regarding gene product function, particularly in the context
of environmental exposure.
Also, the genotypes of animals, as in genetically manipulated animal models (e.g. gene
knockout and transgenic mice), have been used effectively for understanding the
metabolism of toxicants mediated by cytochrome P450 (CYP) isozymes, in order to further
elucidate mechanisms of toxicity. For example, MS-based approaches were able to
distinguish between the metabolic profiles for Cyp2e1-null, CYP1A2-humanized, and wild-
type mice after exposure to the ubiquitous dietary carcinogen 2-amino-1-methyl-6-
phenylimidazo[4,5-b]pyridine (Chen et al., 2007) or the hepatotoxic agent acetaminophen
(Chen et al., 2008). As a result, the metabolomics approach could be used to identify
mechanistic changes stemming from genetic differences.

5.3 Organ-specific effects


Metabolomics approach has been utilized in identifying specific profiles that are altered in
different organs in response to toxicant injury. The following represents a brief discussion of
three select organ toxicities associated with exposure to a given chemical or mixture of
Metabolomics Approach for Hazard Identification in
Human Health Assessment of Environmental Chemicals 353

chemicals. The extent of the metabolomic datasets varies for each type of organ toxicity;
however, discussion focuses on how metabolomic investigations have contributed to some
understanding of mechanisms of toxicity. Some of these metabolic changes are specific but
many are nonspecific for the selected organ toxicities.

5.3.1 Hepatotoxicity (Carbon tetrachloride)


Carbon tetrachloride (CCl4) is a well-studied hepatotoxicant and carcinogen that has been
used as a fumigant (Manibusan et al., 2007). The mechanisms underlying CCl4 toxic
responses are fairly well understood. In rats, CCl4 exposure results in mild to severe
centrilobular necrosis of the liver with elevation of serum alanine aminotransferase (ALT)
and aspartate aminotransferase (AST) activities suggestive of hepatotoxicity. In addition,
CCl4 induces lipid peroxidation, oxidative stress, and regenerative proliferation, eventually
leading to hepatocarcinogenesis. The following brief discussion focuses on how
metabolomics has been used to identify metabolites associated with these known
mechanisms of toxic response.
Metabolomic analyses have identified several aldehydes (e.g. formaldehyde, acetaldehyde,
propanal, butanal, pentanal, hexanal and malondialdehyde) excreted in urine (De Zwart et
al., 1997) or in exhaled breath (Gee et al., 1981) from rodents dosed with CCl4. Similarly,
short-chain hydrocarbons (e.g. pentane and ethane) generated during lipid peroxidation
have been reported in the breath of humans suggestive of CCl4-induced oxidative stress
(Hwang & Kim, 2007). These reactive aldehydes arising from lipid peroxidation have been
shown to be quenched by reduced glutathione (GSH), causing GSH depletion which leads to
cirrhosis in the liver of rats (Cabre et al., 2000). Further, GSH depletion causes a perturbation
of prooxidant-antioxidant balance eventually leading to oxidative stress (Ichi et al., 2009).
Metabolomic studies have provided support that oxidative stress induced by CCl4 generates
reactive oxygen species (ROS) capable of causing DNA damage as shown by an increase in
8-oxodeoxyguanosine (8-oxodG) and malondialdehyde-deoxyguanosine (M1G) adducts
(Beddowes et al., 2003). Also, lipid peroxidation products, such as 4-hydroxynonenal, can
form etheno adducts, which are capable of inducing mutations in critical genes through
base-pair substitutions (Barbin, 2000; Chung et al., 1999).
Further, CCl4-induced oxidative stress inhibits energy metabolism as observed by changes
in levels of Krebs cycle intermediates (i.e. citric acid, 2-oxoglutarate, and succinate), which
has an overall effect on glutathione metabolism eventually leading to hepatotoxicity (Huang
et al., 2008; Kim et al., 2008; Lin et al., 2009; Robertson et al., 2000). These earlier cellular
perturbations further affect Ca+2 homeostasis, cause protein and phospholipid degradation,
and potentially lead to cytotoxicity (Manibusan et al., 2007), apoptosis and necrosis of the
liver tissue (Sun et al., 2001). Furthermore, membrane phospholipid degradation leads to the
release of high levels of o-phosphoethanolamine, a well-known biomarker of cytotoxicity
that can be detected using a metabolomic approach (Wang et al., 2009).
CCl4 toxicity also leads to an inflammatory response characterized by the release of
arachidonic acid, which is the precursor for a cascade of events leading to prostaglandin and
leukotriene synthesis involving cyclooxygenases and lipoxygenases, respectively (Basu,
2003), and other newer inflammatory biomarkers, such as resolvins, protectins, and
maresins (Liu et al., 2009; Serhan, 2009). Following cytotoxicity, liver tissue undergoes
354 Metabolomics

regenerative proliferation and activation of the urea cycle characterized by polyamine


biosynthesis, the latter being a hallmark of cellular proliferation and differentiation (Heby,
1981). Detection of increased levels of urea cycle metabolites such as putrescine, ornithine,
spermine, and spermidine (Pegg et al., 1981) in CCl4–exposed rats by metabolomics
approach gives an early indication of regenerative cell proliferation, which has been
detected at a later stage of disease progression using conventional toxicity assays.
Overall, metabolomics studies have identified important biomarkers for CCl4–induced
toxicity including: aldehydes (lipid peroxidation); GSH/GSSG ratio (oxidative stress); 8-
oxodG, M1G, and etheno adducts (genotoxicity); o-phosphoethanolamine (cytotoxicity);
arachidonic acid and prostaglandins (inflammation); and polyamines (regenerative cell
proliferation), which are also observed in conventional assays (Vulimiri et al., 2011). This
brief discussion further supports the use of metabolomics for investigating toxicity
associated with environmentally related chemicals.

5.3.2 Neurotoxicity (Methylmercury)


Methylmercury is a chemical that was originally used as a fungicide for agricultural
purposes in the early 20th century. In one of the earliest studies of methylmercury toxicity,
occupational exposure to this chemical resulted in detriments in neurological functions in
factory workers and changes in brain morphology with specificity to the cerebellar area.
Additionally, in one of the most noted disasters in the 20th century, methylmercury exposure
through consumption of fish was determined to be the causative agent for producing
neurological deficits in fishermen and neurodevelopmental abnormalities in children in
Minamata Bay, Japan (Ekino et al., 2007).
Traditional approaches in hazard characterization have concluded that the most sensitive
(critical) effect observed from methylmercury exposure is neurobehavioral changes in
children [as reviewed in (U.S. EPA, 2005) and Agency for Toxic Substances and Disease
Registry (ATSDR, 1999)]. An uncertainty that was highlighted in both of these human health
assessment evaluations was that of pharmacodynamic variability in the human population.
One approach to address pharmacodynamic variability is to have an understanding of the
key mechanism(s) that results in the sensitive effect. There have been several reviews on the
neurological mechanisms associated with methylmercury (Ekino et al., 2007; Myers et al.,
2009). One of the primary mechanisms associated with methylmercury neurotoxicity in both
the adult and developing brain is cytotoxicity. Methylmercury has been demonstrated to
induce oxidative stress in the nervous system. Additionally, in vitro studies have reported
that methylmercury treatment results in apoptosis in glial cells, cerebellar granule cells, and
in neuronal cell lines such as differentiated PC12 cells and neuroblastoma cells.
Methylmercury exposure in rats has also resulted in apoptosis in the cerebellum as well as
loss of astrocytes in monkeys. The cell death pathway for methylmercury has been highly
characterized as reviewed recently (Ceccatelli et al., 2010). In summary, several studies have
demonstrated that methylmercury induces caspase-dependent apoptosis in primary
neuronal cell cultures and cell lines.
Metabolomic analysis for methylmercury was conducted using an in vitro methodology
proposed by the European Commission Joint Research Centre in 2008 (van Vliet et al., 2008).
Dynamic reaggregating neuronal cultures prepared from 16-day old rat fetal telencephalon
Metabolomics Approach for Hazard Identification in
Human Health Assessment of Environmental Chemicals 355

were used for this analysis. The identified putative biomarkers were γ-aminobutyric acid
(GABA), choline, glutamine, creatine, and spermine. The authors were able to speculate
potential mechanisms of methylmercury from results of traditional biochemical assays
(Eskes et al., 2002; Monnet-Tschudi et al., 1996) where the enzymatic activities of neuronal
enzymes including glutamine synthesis, choline acetyltransferase, and glutamic acid
decarboxylase were reported to be significantly decreased. In essence, the decreased levels
of GABA and choline in the in vitro metabolomic study may help to explain these decreases
in enzymatic activity which then leads to neuron-specific toxicity. This information
correlates to the observed increases in apoptotic cell death associated with methylmercury.
Similarly, the increased creatine levels that were reported from methylmercury treatment
were correlated to gliosis (proliferation of astrocytes in the central nervous system) by the
authors. Creatine is generally linked to brain osmoregulation (Bothwell et al., 2001) and
increased creatine levels would lead to increased activity of glial cells that could result in
gliosis.
Thus, a very limited metabolomic dataset for methylmercury adds to the mechanistic profile
of this compound and helps establish a temporality of the key events leading to cytotoxicity
in the brain. In the case of using the metabolomic data for methylmercury, the observed
changes in the neuronal metabolites provide supportive, early evidence of later stage events
leading to brain cytotoxicity. Metabolomic data from in vivo studies or from incidental
human exposure would demonstrate any pharmacodynamic differences and if so, how such
differences could be quantified through the available data rather than the default use of
uncertainty factors.

5.3.3 Pulmonary toxicity (Cigarette smoke)


Cigarette smoking is one of the major etiological reasons for pulmonary toxicity and lung
cancer. Cigarette smoke contain at least 4000 chemicals from different chemical classes, of
which at least 60 are well-established carcinogens in experimental animals (Hecht, 2006).
Cigarette smoke has been shown to induce oxidative stress and an inflammatory response in
lungs, and chronic exposure is known to cause cancer. While cigarette smoke is most
deleterious to the smoker, environmental or passive exposure to smoke (i.e. second-hand
smoke) can lead to adverse health effects to bystanders (U.S. EPA, 1992). Using the
metabolomics approach in A549 human lung epithelial cells it has been shown that several
biochemical pathways are altered by either the whole smoke (WS) or its component phases
i.e. wet total particulate matter (WTPM) or gas/vapor phase (GVP) (Vulimiri et al., 2009).
Exposures to the different phases lead to unique biochemical alterations in A549 cells. For
example, WTPM exposure resulted in changes in the Krebs cycle and urea cycle metabolites,
whereas GVP exposure resulted in alterations in hexosamines and in lipid metabolism.
These investigators found that exposure to cigarette smoke was associated with
perturbations in the metabolism of glutathione, amino acids, lipids, and nucleotides.
Alterations were also seen in the urea cycle, Krebs cycle, and the production of polyamines
and cellular cofactors.
Oxidative stress is an important consequence associated with cigarette smoke. The oxidants
produced or found in cigarette smoke can react with and damage cellular macromolecules
including proteins, DNA, and lipid membranes. Additionally, these oxidants can act as
signaling molecules that can influence cell proliferation (Faux et al., 2009). However, cells
356 Metabolomics

contain antioxidant defenses, such as GSH to prevent oxidative damage (Rahman &
MacNee, 1999). The metabolomics approach has indeed found changes in metabolites
associated with these effects of cigarette smoke. Predominant changes in metabolites from
exposure to WS included glutathione and -glutamylglutamine, which showed 51 and 13-
fold increases compared to control cells. The increased levels of metabolites within the
glutathione pathway (i.e. glutathione and -glutamylglutamine) suggest a protective
response against oxidative stress, which can result from WS. These data correlate with
human microarray data that demonstrated an antioxidant response to cigarette smoke
through the induction of genes associated with glutathione metabolism (Spira et al., 2004).
Further, Vulimiri and colleagues (Vulimiri et al., 2009) observed increased levels (16.4-fold)
of o-phosphoethanolamine, a marker of phospholipid degradation that may indicate cell
membrane damage. Additional observations included increased arachidonic acid levels that
may suggest an inflammatory response and markers (e.g. putrescine) of cell proliferation.
Conversely, there was a statistically significant decrease in glutathione levels from exposure
to WTPM and GVP when compared to controls. Predominant metabolite changes in these
phases were acetylcarnitine (4.5-fold increase) and palmitoleate (5.2-fold increase) for
WTPM and GVP, respectively, both of which indicated changes in lipid metabolism
(Vulimiri et al., 2009). In summary, these authors (Vulimiri et al., 2009) demonstrated how
metabolomics could differentiate metabolic responses caused by exposure to complex
mixtures (i.e. cigarette smoke) and also provided empirical data for metabolic changes for
known markers of toxicity (e.g. decreased GSH, membrane damage) associated with
exposure to cigarette smoke.

6. Integrating “omics” data into risk assessment


The field of toxicology is advancing with several of the high throughput screening
techniques taking major roles in understanding toxicity pathways. More specifically
toxicogenomics and proteomics have contributed to identifying the mechanisms of toxicity
pathways (Burchiel et al., 2001; Heijne et al., 2005). Integration of data from different
“omics” approaches would help clarify the mode of action of xenobiotics especially at low
dose levels, which are relevant to environmental exposures. Thus, combining “omic”
techniques such as toxicogenomics, transcriptomics, proteomics, and metabolomics
promises to provide a robust understanding of biological responses to xenobiotics. Under
this paradigm, effects on genes and their downstream products, namely proteins and the
metabolites produced by proteins, can be assessed together. Researchers have utilized
diverse “omics” approaches to understand the mode(s) of action or mechanism of different
toxicity pathways in various experimental models. Some studies have indeed shown a good
correlation between the genomics and metabolomics approaches. For example,
metabolomics approach has demonstrated alterations in the pathways associated with lipid
peroxidation, DNA damage and repair, and cell proliferation in rats exposed to CCl4 which
was consistent with the expression of transcripts associated with steatosis/fibrosis that were
specific to cell injury and regeneration (Chung et al., 2005a; Chung et al., 2005b;
Fountoulakis et al., 2002). Additionally, metabolomics has identified biochemical alterations
in pathways associated with oxidative stress, inflammation, cell proliferation, and
cytotoxicity in human lung epithelial cells exposed to cigarette smoke (Vulimiri et al., 2009).
These observations are supported by data from genomic (Harvey et al., 2007; Meng et al.,
Metabolomics Approach for Hazard Identification in
Human Health Assessment of Environmental Chemicals 357

2006; Spira et al., 2004) and proteomic (Carter et al., 2011; Kelsen et al., 2008; Zhang et al.,
2008) investigations that also detected markers associated with the pathways affected by
exposure to cigarette smoke.
Conversely, the integration of “omics” technologies has also demonstrated some limitations.
For example, a study by Steiling and co-workers found a discrepancy between genomic and
proteomic data. Specifically, gel electrophoresis followed by LC-MS analysis identified 41
proteins whose expression would not have been detected by microarray analysis (Steiling et
al., 2009). Such data highlight the importance of assessing more downstream markers (i.e.
proteins or metabolites) that may provide a more accurate understanding of the biological
responses to chemical exposures.

7. Shift in the risk assessment paradigm from apical endpoints to


biochemical perturbations
Historically, human health risk assessment relied on identifying apical endpoints, defined as
“observable outcomes in a whole organism, such as a clinical sign or pathologic state,
indicative of a disease state that can result from exposure to a toxicant”, such as birth
defects, neurologic deficits, and tumor number that are obtained from high-dose animal
bioassays (Krewski et al., 2010). However, the mechanistic relevance of these data can be
tenuous considering the need for interspecies extrapolation and that environmental
exposures may be orders of magnitude lower. Recently, the NRC report on “Toxicity Testing
in the 21st Century: A Vision and Strategy” suggested a fundamental change in the risk
assessment paradigm where the reliance on apical endpoints of toxicity would be replaced
in favor of in vitro toxicity testing for identifying perturbations in biochemical pathways.
Such an approach would employ cell lines, particularly human-based lines, and high
throughput screening assays (e.g. genomics, proteomics) with computational toxicology
methods. As a result, this approach would also lead to an overall increase in the efficiency of
toxicity testing and decrease in animal usage [as reviewed in (Krewski et al., 2010)]. As
pointed out by the NRC committee, the use of these low-dose, human-based in vitro systems
would negate concerns associated with high-dose, animal data. Since metabolomics can
identify early perturbations in biochemical pathways, this technology is poised to become
an important element of this proposed risk assessment paradigm.

8. Advantages of metabolomics approach for environmental chemical


assessment
Metabolomics approach offers several advantages for understanding the mechanisms of
toxicity of environmental chemicals and informing human health assessments. One
advantage of the metabolomics approach is the relatively non-invasive (e.g. urine samples)
nature of this technique. In this context, samples from humans subjected to an incidental
environmental exposure can be easily collected, analyzed, and correlated to metabolomic
profiles from animal studies in order to identify critical effects from the exposure. Aside
from quantitative differences, metabolic pathways are evolutionally conserved across
different species; metabolomics data can be qualitatively extrapolated or interpreted at the
molecular level among and between species. Unlike genomics and proteomics,
metabolomics databases offer information on the structural, physicochemical,
358 Metabolomics

pharmacological and spectral profiles as well as biological functions of metabolites (Go,


2010). From a technical standpoint, sample preparation is relatively minimal for
metabolomic analyses compared to genomics and proteomics approaches. Also, since
metabolomics allows for the a priori selection of metabolites, the investigator has better
control over the number of standards that need to be procured or synthesized for analysis.
A survey of the available literature already demonstrates the value of using
metabolomics for elucidating the mode of action of environmental chemicals. Since a
number of matrices are amenable to metabolomic analysis, various systems including in
vitro and in vivo models can be employed to identify metabolic perturbations associated
with exposure to environmental chemicals. Some of the mammalian in vitro systems
involved exposing A549 human lung carcinoma cells to cigarette smoke (Vulimiri et al.,
2009), human embryonic stem cells to various teratogens (West et al., 2010), and
reaggregating neuronal cultures to methylmercury (van Vliet et al., 2008). From in vivo
studies, intact organs from exposed experimental animals have been evaluated to include
the analysis of intact lung after intratracheal instillation of silica dust (Hu et al., 2008) as
well as liver and lung after exposure to 1-nitronaphthalene (Azmi et al., 2005). Also,
metabolomics has been able to demonstrate different metabolic responses between
mixtures of environmental chemicals. For example, in assessing the inflammatory
response, Schmelzer et al (2006) found unique lipid profiles between rats exposed to 1-
nitronaphthalene and those exposed to a mixture of 1-nitronaphthalene and ozone. As
highlighted earlier in this chapter, metabolomics approach has been shown to
differentiate the metabolic changes attributed to the complex mixtures of whole smoke
versus its two constituent physical phases (Vulimiri et al., 2009).

9. Limitations of metabolomics research


Regardless of whether a chemical being investigated is a pharmaceutical or environmental
pollutant, a critique of metabolomics has been the detection of metabolites that appear to
be changed independent of the toxicant, its mode of action, and target tissue. Such
common metabolites were previously termed the “usual suspects” by Robertson and often
include 2-oxoglutarate, citrate, succinate, and trimethyl amine/trimethyl amine oxide
(Robertson, 2005). From a data interpretation standpoint, these “usual suspects” may
confound the identification of metabolites that have true value as specific markers of
organ toxicity by skewing results from pattern separation analyses (Connor et al., 2004;
Robertson, 2005). Connor et al (2004) found that urinary metabolite changes, including
many of the “usual suspect” metabolites, can be associated with systemic or secondary
toxicity effects, namely reductions in food intake and body weight. Additionally, the
authors suggest that such metabolites may still inform mechanisms of toxicity when put
into the context of diet and weight change. Conversely, the authors did identify exposures
where common metabolites were specific for toxicant exposure. For example, succinate
and 2-oxoglutarate were specifically associated with exposure to the hepatotoxicant α-
naphthylisothiocyanate, rather than resulting from secondary toxicity (Connor et al.,
2004). Research by other investigators has identified metabolites as novel biomarkers
associated with specific diseases and for specific toxicities. For example, 5-oxoproline has
been demonstrated to be a specific marker of bromobenzene-induced hepatotoxicity
(Waters et al., 2006).
Metabolomics Approach for Hazard Identification in
Human Health Assessment of Environmental Chemicals 359

10. Future directions/research needs


A goal of this chapter has been to highlight how metabolomics approach can be used to
better understand the toxicity of environmental chemicals, with a particular focus on hazard
identification and mode of action. However, due to its relative infancy compared to
conventional toxicity assays and other “omic” technologies, metabolomic information for
environmental chemicals is sparse. As interest in metabolomics increases and as this
approach becomes more accessible to researchers focused on environmental chemicals, this
database will grow. A contemporary issue regarding xenobiotics is the influence of the
microbiome on immunity, metabolism, and human health [reviewed in (Han et al., 2010)].
This microbiome involves the sum of all the microorganisms (e.g. bacteria) that internally
and externally reside on an individual or animal. Of these microorganisms, the gut
microbiota has been shown to influence the metabolism of several xenobiotics. Accordingly,
the microbiome is likely to be important in the toxicity of environmental chemicals and
pertinent to human health assessments (Betts, 2011). Thus, the influence of the microbiome
is an important aspect of chemical toxicity that should be further studied using
metabolomics.

11. Conclusions
Metabolomics is an emerging medium-to-high throughput technique which measures the
endogenous biochemicals affecting different metabolic pathways and can be useful in
characterizing the hazards of environmental chemicals. Identifying metabolic perturbations
caused in mammalian cell systems following chemical exposure helps in elucidating the
predominant toxicity pathways. Some of the advantages of using metabolomic data for
hazard identification—one of the key steps in risk assessment—include the ability to inform
gender, genetic, and organ-specific effects in a relatively expedient manner. As briefly
discussed, metabolomics can identify early biochemical perturbations associated with
toxicity in the hepatic, nervous, and pulmonary systems caused by selected environmental
chemicals. As surveyed, various research systems using metabolomics demonstrate how
metabolomic data could be used for hazard identification and mode of action
characterization for environmental chemicals. Overall, metabolomics represents an
opportunity to develop a better understanding of the toxicity of environmental chemicals
and could further impact the human health assessment of these chemicals.

12. Disclaimer
The views expressed in this chapter are those of the authors and do not necessarily represent
the views or policies of the U.S. Environmental Protection Agency.

13. References
ATSDR (1999). Toxicological profile for mercury, Agency for Toxic Substances and Disease
Registry, Atlanta, GA, Available from:
http://www.atsdr.cdc.gov/toxprofiles/tp46.pdf
Azmi, J.; Connelly, J.; Holmes, E.; Nicholson, J.K.; Shore, R.F. & Griffin, J.L. (2005).
Characterization of the biochemical effects of 1-nitronaphthalene in rats using
360 Metabolomics

global metabolic profiling by NMR spectroscopy and pattern recognition,


Biomarkers, Vol. 10, No. 6, pp. 401-16.
Barbin, A. (2000). Etheno-adduct-forming chemicals: from mutagenicity testing to tumor
mutation spectra, Mutat Res, Vol. 462, No. 2-3, pp. 55-69.
Basu, S. (2003). Carbon tetrachloride-induced lipid peroxidation: eicosanoid formation and
their regulation by antioxidant nutrients, Toxicology, Vol. 189, No. 1-2, pp. 113-27.
Beddowes, E.J.; Faux, S.P. & Chipman, J.K. (2003). Chloroform, carbon tetrachloride and
glutathione depletion induce secondary genotoxicity in liver cells via oxidative
stress, Toxicology, Vol. 187, No. 2-3, pp. 101-15.
Betts, K.S. (2011). A study in balance: how microbiomes are changing the shape of
environmental health, Environ Health Perspect, Vol. 119, No. 8, pp. a340-6.
Bothwell, J.H.; Rae, C.; Dixon, R.M.; Styles, P. & Bhakoo, K.K. (2001). Hypo-osmotic
swelling-activated release of organic osmolytes in brain slices: implications for
brain oedema in vivo, J Neurochem, Vol. 77, No. 6, pp. 1632-40.
Boverhof, D.R. & Zacharewski, T.R. (2006). Toxicogenomics in risk assessment: applications
and needs, Toxicol Sci, Vol. 89, No. 2, pp. 352-60.
Burchiel, S.W.; Knall, C.M.; Davis, J.W., 2nd; Paules, R.S.; Boggs, S.E. & Afshari, C.A. (2001).
Analysis of genetic and epigenetic mechanisms of toxicity: potential roles of
toxicogenomics and proteomics in toxicology, Toxicol Sci, Vol. 59, No. 2, pp. 193-5.
Cabre, M.; Camps, J.; Paternain, J.L.; Ferre, N. & Joven, J. (2000). Time-course of changes in
hepatic lipid peroxidation and glutathione metabolism in rats with carbon
tetrachloride-induced cirrhosis, Clin Exp Pharmacol Physiol, Vol. 27, No. 9, pp. 694-9.
Carter, C.A.; Misra, M. & Pelech, S. (2011). Proteomic Analyses of Lung Lysates from Short-
Term Exposure of Fischer 344 Rats to Cigarette Smoke, J Proteome Res, Vol. (In
Press), No.
Ceccatelli, S.; Dare, E. & Moors, M. (2010). Methylmercury-induced neurotoxicity and
apoptosis, Chem Biol Interact, Vol. 188, No. 2, pp. 301-8.
Chen, C.; Krausz, K.W.; Idle, J.R. & Gonzalez, F.J. (2008). Identification of novel toxicity-
associated metabolites by metabolomics and mass isotopomer analysis of
acetaminophen metabolism in wild-type and Cyp2e1-null mice, J Biol Chem, Vol.
283, No. 8, pp. 4543-59.
Chen, C.; Ma, X.; Malfatti, M.A.; Krausz, K.W.; Kimura, S.; Felton, J.S.; Idle, J.R. & Gonzalez,
F.J. (2007). A comprehensive investigation of 2-amino-1-methyl-6-
phenylimidazo[4,5-b]pyridine (PhIP) metabolism in the mouse using a multivariate
data analysis approach, Chem Res Toxicol, Vol. 20, No. 3, pp. 531-42.
Chung, F.L.; Nath, R.G.; Nagao, M.; Nishikawa, A.; Zhou, G.D. & Randerath, K. (1999).
Endogenous formation and significance of 1,N2-propanodeoxyguanosine adducts,
Mutat Res, Vol. 424, No. 1-2, pp. 71-81.
Chung, H.; Hong, D.P.; Jung, J.Y.; Kim, H.J.; Jang, K.S.; Sheen, Y.Y.; Ahn, J.I.; Lee, Y.S. &
Kong, G. (2005a). Comprehensive analysis of differential gene expression profiles
on carbon tetrachloride-induced rat liver injury and regeneration, Toxicol Appl
Pharmacol, Vol. 206, No. 1, pp. 27-42.
Chung, H.; Hong, D.P.; Kim, H.J.; Jang, K.S.; Shin, D.M.; Ahn, J.I.; Lee, Y.S. & Kong, G.
(2005b). Differential gene expression profiles in the steatosis/fibrosis model of rat
liver by chronic administration of carbon tetrachloride, Toxicol Appl Pharmacol, Vol.
208, No. 3, pp. 242-54.
Connor, S.C.; Wu, W.; Sweatman, B.C.; Manini, J.; Haselden, J.N.; Crowther, D.J. &
Waterfield, C.J. (2004). Effects of feeding and body weight loss on the 1H-NMR-
Metabolomics Approach for Hazard Identification in
Human Health Assessment of Environmental Chemicals 361

based urine metabolic profiles of male Wistar Han rats: implications for biomarker
discovery, Biomarkers, Vol. 9, No. 2, pp. 156-79.
De Zwart, L.L.; Venhorst, J.; Groot, M.; Commandeur, J.N.; Hermanns, R.C.; Meerman, J.H.;
Van Baar, B.L. & Vermeulen, N.P. (1997). Simultaneous determination of eight lipid
peroxidation degradation products in urine of rats treated with carbon
tetrachloride using gas chromatography with electron-capture detection, J
Chromatogr B Biomed Sci Appl, Vol. 694, No. 2, pp. 277-87.
Ekino, S.; Susa, M.; Ninomiya, T.; Imamura, K. & Kitamura, T. (2007). Minamata disease
revisited: an update on the acute and chronic manifestations of methyl mercury
poisoning, J Neurol Sci, Vol. 262, No. 1-2, pp. 131-44.
Eskes, C.; Honegger, P.; Juillerat-Jeanneret, L. & Monnet-Tschudi, F. (2002). Microglial
reaction induced by noncytotoxic methylmercury treatment leads to
neuroprotection via interactions with astrocytes and IL-6 release, Glia, Vol. 37, No.
1, pp. 43-52.
Faux, S.P.; Tai, T.; Thorne, D.; Xu, Y.; Breheny, D. & Gaca, M. (2009). The role of oxidative
stress in the biological responses of lung epithelial cells to cigarette smoke,
Biomarkers, Vol. 14 Suppl 1, No. 90-6.
Fountoulakis, M.; de Vera, M.C.; Crameri, F.; Boess, F.; Gasser, R.; Albertini, S. & Suter, L.
(2002). Modulation of gene and protein expression by carbon tetrachloride in the
rat liver, Toxicol Appl Pharmacol, Vol. 183, No. 1, pp. 71-80.
Gee, D.L.; Bechtold, M.M. & Tappel, A.L. (1981). Carbon tetrachloride-induced lipid
peroxidation: simultaneous in vivo measurements of pentane and chloroform
exhaled by the rat, Toxicol Lett, Vol. 8, No. 6, pp. 299-306.
Gieger, C.; Geistlinger, L.; Altmaier, E.; Hrabe de Angelis, M.; Kronenberg, F.; Meitinger, T.;
Mewes, H.W.; Wichmann, H.E.; Weinberger, K.M.; Adamski, J.; Illig, T. & Suhre, K.
(2008). Genetics meets metabolomics: a genome-wide association study of
metabolite profiles in human serum, PLoS Genet, Vol. 4, No. 11, pp. e1000282.
Ginsberg, G.; Smolenski, S.; Neafsey, P.; Hattis, D.; Walker, K.; Guyton, K.Z.; Johns, D.O. &
Sonawane, B. (2009). The influence of genetic polymorphisms on population
variability in six xenobiotic-metabolizing enzymes, J Toxicol Environ Health B Crit
Rev, Vol. 12, No. 5-6, pp. 307-33.
Go, E.P. (2010). Database resources in metabolomics: an overview, J Neuroimmune Pharmacol,
Vol. 5, No. 1, pp. 18-30.
Guyton, K.Z.; Kyle, A.D.; Aubrecht, J.; Cogliano, V.J.; Eastmond, D.A.; Jackson, M.; Keshava,
N.; Sandy, M.S.; Sonawane, B.; Zhang, L.; Waters, M.D. & Smith, M.T. (2009).
Improving prediction of chemical carcinogenicity by considering multiple
mechanisms and applying toxicogenomic approaches, Mutat Res, Vol. 681, No. 2-3,
pp. 230-40.
Han, J.; Antunes, L.C.; Finlay, B.B. & Borchers, C.H. (2010). Metabolomics: towards
understanding host-microbe interactions, Future Microbiol, Vol. 5, No. 2, pp. 153-61.
Harvey, B.G.; Heguy, A.; Leopold, P.L.; Carolan, B.J.; Ferris, B. & Crystal, R.G. (2007).
Modification of gene expression of the small airway epithelium in response to
cigarette smoking, J Mol Med (Berl), Vol. 85, No. 1, pp. 39-53.
Heby, O. (1981). Role of polyamines in the control of cell proliferation and differentiation,
Differentiation, Vol. 19, No. 1, pp. 1-20.
Hecht, S.S. (2006). Cigarette smoking: cancer risks, carcinogens, and mechanisms,
Langenbecks Arch Surg, Vol. 391, No. 6, pp. 603-13.
362 Metabolomics

Heijne, W.H.; Kienhuis, A.S.; van Ommen, B.; Stierum, R.H. & Groten, J.P. (2005). Systems
toxicology: applications of toxicogenomics, transcriptomics, proteomics and
metabolomics in toxicology, Expert Rev Proteomics, Vol. 2, No. 5, pp. 767-80.
Hu, J.Z.; Rommereim, D.N.; Minard, K.R.; Woodstock, A.; Harrer, B.J.; Wind, R.A.; Phipps,
R.P. & Sime, P.J. (2008). Metabolomics in lung inflammation:a high-resolution (1)h
NMR study of mice exposedto silica dust, Toxicol Mech Methods, Vol. 18, No. 5, pp.
385-98.
Huang, X.; Shao, L.; Gong, Y.; Mao, Y.; Liu, C.; Qu, H. & Cheng, Y. (2008). A metabonomic
characterization of CCl4-induced acute liver failure using partial least square
regression based on the GC/MS metabolic profiles of plasma in mice, J Chromatogr
B Analyt Technol Biomed Life Sci, Vol. 870, No. 2, pp. 178-85.
Hwang, E.S. & Kim, G.H. (2007). Biomarkers for oxidative stress status of DNA, lipids, and
proteins in vitro and in vivo cancer research, Toxicology, Vol. 229, No. 1-2, pp. 1-10.
Ichi, I.; Kamikawa, C.; Nakagawa, T.; Kobayashi, K.; Kataoka, R.; Nagata, E.; Kitamura, Y.;
Nakazaki, C.; Matsura, T. & Kojo, S. (2009). Neutral sphingomyelinase-induced
ceramide accumulation by oxidative stress during carbon tetrachloride
intoxication, Toxicology, Vol. 261, No. 1-2, pp. 33-40.
Idle, J.R. & Gonzalez, F.J. (2007). Metabolomics, Cell Metab, Vol. 6, No. 5, pp. 348-51.
Kelada, S.N.; Eaton, D.L.; Wang, S.S.; Rothman, N.R. & Khoury, M.J. (2003). The role of
genetic polymorphisms in environmental health, Environ Health Perspect, Vol. 111,
No. 8, pp. 1055-64.
Kelsen, S.G.; Duan, X.; Ji, R.; Perez, O.; Liu, C. & Merali, S. (2008). Cigarette smoke induces
an unfolded protein response in the human lung: a proteomic approach, Am J
Respir Cell Mol Biol, Vol. 38, No. 5, pp. 541-50.
Kim, K.-B.; Chung, M.W.; Um, S.Y.; Oh, J.S.; Kim, S.H.; Na, M.A.; Oh, H.Y.; Cho, W.-S. &
Choi, K.H. (2008). Metabolomics and biomarker discovery: NMR spectral data of
urine and hepatotoxicity by carbon tetrachloride, acetaminophen, and D-
galactosamine in rats, Metabolomics, Vol. 4, No. 377-392.
Krewski, D.; Acosta, D., Jr.; Andersen, M.; Anderson, H.; Bailar, J.C., 3rd; Boekelheide, K.;
Brent, R.; Charnley, G.; Cheung, V.G.; Green, S., Jr.; Kelsey, K.T.; Kerkvliet, N.I.; Li,
A.A.; McCray, L.; Meyer, O.; Patterson, R.D.; Pennie, W.; Scala, R.A.; Solomon,
G.M.; Stephens, M.; Yager, J. & Zeise, L. (2010). Toxicity testing in the 21st century:
a vision and a strategy, J Toxicol Environ Health B Crit Rev, Vol. 13, No. 2-4, pp. 51-
138.
Lin, Y.; Si, D.; Zhang, Z. & Liu, C. (2009). An integrated metabonomic method for profiling
of metabolic changes in carbon tetrachloride induced rat urine, Toxicology, Vol. 256,
No. 3, pp. 191-200.
Liu, J.Y.; Tsai, H.J.; Hwang, S.H.; Jones, P.D.; Morisseau, C. & Hammock, B.D. (2009).
Pharmacokinetic optimization of four soluble epoxide hydrolase inhibitors for use
in a murine model of inflammation, Br J Pharmacol, Vol. 156, No. 2, pp. 284-96.
Manibusan, M.K.; Odin, M. & Eastmond, D.A. (2007). Postulated carbon tetrachloride mode
of action: a review, J Environ Sci Health Part C, Vol. 25, No. 3, pp. 185-209.
Meng, Q.R.; Gideon, K.M.; Harbo, S.J.; Renne, R.A.; Lee, M.K.; Brys, A.M. & Jones, R. (2006).
Gene expression profiling in lung tissues from mice exposed to cigarette smoke,
lipopolysaccharide, or smoke plus lipopolysaccharide by inhalation, Inhal Toxicol,
Vol. 18, No. 8, pp. 555-68.
Metabolomics Approach for Hazard Identification in
Human Health Assessment of Environmental Chemicals 363

Monnet-Tschudi, F.; Zurich, M.G. & Honegger, P. (1996). Comparison of the developmental
effects of two mercury compounds on glial cells and neurons in aggregate cultures
of rat telencephalon, Brain Res, Vol. 741, No. 1-2, pp. 52-9.
Mortensen, H.M. & Euling, S.Y. (2011). Integrating mechanistic and polymorphism data to
characterize human genetic susceptibility for environmental chemical risk
assessment in the 21st century, Toxicol Appl Pharmacol, Vol. No. (In Press).
Myers, G.J.; Thurston, S.W.; Pearson, A.T.; Davidson, P.W.; Cox, C.; Shamlaye, C.F.;
Cernichiari, E. & Clarkson, T.W. (2009). Postnatal exposure to methyl mercury from
fish consumption: a review and new data from the Seychelles Child Development
Study, Neurotoxicology, Vol. 30, No. 3, pp. 338-49.
NRC (1983). Risk assessment in the federal government: Managing the process, National
Academies Press, Washington, DC. Available from:
http://www.nap.edu/openbook.php?isbn=0309033497
NRC (1994). Science and judgment in risk assessment, National Academies Press, Washington,
DC. Available from: http://www.nap.edu/catalog/2125.html
Pegg, A.E.; Matsui, I.; Seely, J.E.; Pritchard, M.L. & Poso, H. (1981). Formation of putrescine
in rat liver, Med Biol, Vol. 59, No. 5-6, pp. 327-33.
Raamsdonk, L.M.; Teusink, B.; Broadhurst, D.; Zhang, N.; Hayes, A.; Walsh, M.C.; Berden,
J.A.; Brindle, K.M.; Kell, D.B.; Rowland, J.J.; Westerhoff, H.V.; van Dam, K. &
Oliver, S.G. (2001). A functional genomics strategy that uses metabolome data to
reveal the phenotype of silent mutations, Nat Biotechnol, Vol. 19, No. 1, pp. 45-50.
Rahman, I. & MacNee, W. (1999). Lung glutathione and oxidative stress: implications in
cigarette smoke-induced airway disease, Am J Physiol, Vol. 277, No. 6 Pt 1, pp.
L1067-88.
Robertson, D.G. (2005). Metabonomics in toxicology: a review, Toxicol Sci, Vol. 85, No. 2, pp.
809-22.
Robertson, D.G.; Reily, M.D.; Sigler, R.E.; Wells, D.F.; Paterson, D.A. & Braden, T.K. (2000).
Metabonomics: evaluation of nuclear magnetic resonance (NMR) and pattern
recognition technology for rapid in vivo screening of liver and kidney toxicants,
Toxicol Sci, Vol. 57, No. 2, pp. 326-37.
Roux, A.; Lison, D.; Junot, C. & Heilier, J.F. (2011). Applications of liquid chromatography
coupled to mass spectrometry-based metabolomics in clinical chemistry and
toxicology: A review, Clin Biochem, Vol. 44, No. 1, pp. 119-35.
Rubino, F.M.; Pitton, M.; Di Fabio, D. & Colombi, A. (2009). Toward an "omic"
physiopathology of reactive chemicals: thirty years of mass spectrometric study of
the protein adducts with endogenous and xenobiotic compounds, Mass Spectrom
Rev, Vol. 28, No. 5, pp. 725-84.
Schmelzer, K.R.; Wheelock, A.M.; Dettmer, K.; Morin, D. & Hammock, B.D. (2006). The role of
inflammatory mediators in the synergistic toxicity of ozone and 1-nitronaphthalene in
rat airways, Environ Health Perspect, Vol. 114, No. 9, pp. 1354-60.
Serhan, C.N. (2009). Systems approach to inflammation resolution: identification of novel
anti-inflammatory and pro-resolving mediators, J Thromb Haemost, Vol. 7 Suppl 1,
No. 44-8.
Spira, A.; Beane, J.; Pinto-Plata, V.; Kadar, A.; Liu, G.; Shah, V.; Celli, B. & Brody, J.S. (2004).
Gene expression profiling of human lung tissue from smokers with severe
emphysema, Am J Respir Cell Mol Biol, Vol. 31, No. 6, pp. 601-10.
Steiling, K.; Kadar, A.Y.; Bergerat, A.; Flanigon, J.; Sridhar, S.; Shah, V.; Ahmad, Q.R.; Brody,
J.S.; Lenburg, M.E.; Steffen, M. & Spira, A. (2009). Comparison of proteomic and
364 Metabolomics

transcriptomic profiles in the bronchial airway epithelium of current and never


smokers, PLoS One, Vol. 4, No. 4, pp. e5043.
Sun, F.; Hamagawa, E.; Tsutsui, C.; Ono, Y.; Ogiri, Y. & Kojo, S. (2001). Evaluation of
oxidative stress during apoptosis and necrosis caused by carbon tetrachloride in rat
liver, Biochim Biophys Acta, Vol. 1535, No. 2, pp. 186-91.
Szeto, S.S.; Reinke, S.N.; Sykes, B.D. & Lemire, B.D. (2010). Mutations in the Saccharomyces
cerevisiae succinate dehydrogenase result in distinct metabolic phenotypes
revealed through (1)H NMR-based metabolic footprinting, J Proteome Res, Vol. 9,
No. 12, pp. 6729-39.
U.S. EPA (2005). Guidelines for carcinogenic risk assessment, Risk Assessment Forum, U.S.
Environmental Proection Agency, Washington, DC, EPA/630/P-03/001F.
Available from:
http://www.epa.gov/raf/publications/pdfs/CANCER_GUIDELINES_FINAL_3-
25-05.PDF
U.S. EPA (1992). Respiratory health effects of passive smoking: lung cancer and other
disorders, U.S. Environmental Protection Agency, Washington, DC, EPA/600/6-
90/006F. Available from: http://www.epa.gov/ncea/ets/pdfs/acknowl.pdf
U.S. EPA (2001). Water Quality Criterion for the Protection of Human Health:
Methylmercury, U.S. Environmental Protection Agency, Washington, DC, EPA-
823-R-01-001. Available from:
http://water.epa.gov/scitech/swguidance/standards/criteria/aqlife/pollutants/
methylmercury/upload/2009_01_15_criteria_methylmercury_mercury-
criterion.pdf
van Ravenzwaay, B.; Cunha, G.C.; Leibold, E.; Looser, R.; Mellert, W.; Prokoudine, A.; Walk,
T. & Wiemer, J. (2007). The use of metabolomics for the discovery of new
biomarkers of effect, Toxicol Lett, Vol. 172, No. 1-2, pp. 21-8.
van Vliet, E.; Morath, S.; Eskes, C.; Linge, J.; Rappsilber, J.; Honegger, P.; Hartung, T. &
Coecke, S. (2008). A novel in vitro metabolomics approach for neurotoxicity testing,
proof of principle for methyl mercury chloride and caffeine, Neurotoxicology, Vol.
29, No. 1, pp. 1-12.
Vulimiri, S.V.; Berger, A. & Sonawane, B. (2011). The potential of metabolomic approaches
for investigating mode(s) of action of xenobiotics: case study with carbon
tetrachloride, Mutat Res, Vol. 722, No. 2, pp. 147-53.
Vulimiri, S.V.; Misra, M.; Hamm, J.T.; Mitchell, M. & Berger, A. (2009). Effects of mainstream
cigarette smoke on the global metabolome of human lung epithelial cells, Chem Res
Toxicol, Vol. 22, No. 3, pp. 492-503.
Wang, C.; Yang, J. & Nie, J. (2009). Plasma phospholipid metabolic profiling and biomarkers
of rats following radiation exposure based on liquid chromatography-mass
spectrometry technique, Biomed Chromatogr, Vol. 23, No. 10, pp. 1079-85.
Waters, N.J.; Waterfield, C.J.; Farrant, R.D.; Holmes, E. & Nicholson, J.K. (2006). Integrated
metabonomic analysis of bromobenzene-induced hepatotoxicity: novel induction of
5-oxoprolinosis, J Proteome Res, Vol. 5, No. 6, pp. 1448-59.
West, P.R.; Weir, A.M.; Smith, A.M.; Donley, E.L. & Cezar, G.G. (2010). Predicting human
developmental toxicity of pharmaceuticals using human embryonic stem cells and
metabolomics, Toxicol Appl Pharmacol, Vol. 247, No. 1, pp. 18-27.
Zhang, S.; Xu, N.; Nie, J.; Dong, L.; Li, J. & Tong, J. (2008). Proteomic alteration in lung tissue
of rats exposed to cigarette smoke, Toxicol Lett, Vol. 178, No. 3, pp. 191-6.

You might also like